1
|
Qiu Z. Advancements in autism spectrum disorder research --from mechanisms to interventions. Curr Opin Neurobiol 2025; 93:103048. [PMID: 40359648 DOI: 10.1016/j.conb.2025.103048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 04/14/2025] [Accepted: 04/20/2025] [Indexed: 05/15/2025]
Abstract
This review summarizes recent advancements in the research of autism spectrum disorders (ASD), emphasizing genetic underpinnings and their implications for neurodevelopment and cognitive functions. It explores both syndromic and nonsyndromic ASD, highlighting the discovery of critical ASD-related genes and their mechanistic roles as revealed by studies using genetically engineered mouse and non-human primate models. While these models have shed light on the potential of synaptic dysfunction to disrupt brain development, they also underscore the challenges of replicating complex cognitive dysfunctions observed in ASD. Recent successes in gene therapy, particularly through innovative approaches like gene replacement and base editing, offer promising pathways for addressing genetic anomalies in ASD. These therapeutic strategies, underscored by clinical trials and cutting-edge genetic manipulation techniques, pave the way for potential interventions that could profoundly impact ASD management and treatment.
Collapse
Affiliation(s)
- Zilong Qiu
- Department of Neurology, Songjiang Hospital, Songjiang Research Institute, MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Smith M, Dodis GE, Vanderplow AM, Gonzalez S, Rhee Y, Scrogin K, Gogliotti RG. Potentiation of the M 1 muscarinic acetylcholine receptor normalizes neuronal activation patterns and improves apnea severity in Mecp2 +/- mice. Neurobiol Dis 2025; 208:106859. [PMID: 40021095 PMCID: PMC12076212 DOI: 10.1016/j.nbd.2025.106859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/07/2025] [Accepted: 02/24/2025] [Indexed: 03/03/2025] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder that is caused by loss-of-function mutations in the methyl-CpG binding protein 2 (MeCP2) gene. RTT patients experience a myriad of debilitating symptoms, which include respiratory phenotypes that are often associated with lethality. Our previous work established that expression of the M1 muscarinic acetylcholine receptor (mAchR) is decreased in RTT autopsy samples, and that potentiation of the M1 receptor improves apneas in a mouse model of RTT; however, the population of neurons driving this rescue is unclear. Loss of Mecp2 correlates with excessive neuronal activity in cardiorespiratory nuclei. Since M1 is found on cholinergic interneurons, we hypothesized that M1-potentiating compounds decrease apnea frequency by tempering brainstem hyperactivity. To test this, Mecp2+/- and Mecp2+/+ mice were screened for apneas before and after administration of the M1 positive allosteric modulator (PAM) VU0453595 (VU595). Brains from the same mice were then imaged for c-Fos, ChAT, and Syto16 using whole-brain light-sheet microscopy to establish genotype and drug-dependent activation patterns that could be correlated with VU595's efficacy on apneas. The vehicle-treated Mecp2+/- brain exhibited broad hyperactivity when coupled with the phenotypic prescreen, which was significantly decreased by administration of VU595, particularly in regions known to modulate the activity of respiratory nuclei (i.e. hippocampus and striatum). Further, the extent of apnea rescue in each mouse showed a significant positive correlation with c-Fos expression in non-cholinergic neurons in the striatum, thalamus, dentate gyrus, and within the cholinergic neurons of the brainstem. These results indicate that Mecp2+/- mice are prone to hyperactivity in brain regions that regulate respiration, which can be normalized through M1 potentiation.
Collapse
Affiliation(s)
- Mackenzie Smith
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, 2160 S 1st Avenue, Maywood, IL 60153, USA; Edward Hines Jr. VA Hospital, 5000 5th Ave, Hines, IL 60141, USA
| | - Grace E Dodis
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, 2160 S 1st Avenue, Maywood, IL 60153, USA; Edward Hines Jr. VA Hospital, 5000 5th Ave, Hines, IL 60141, USA
| | - Amanda M Vanderplow
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, 2160 S 1st Avenue, Maywood, IL 60153, USA; Edward Hines Jr. VA Hospital, 5000 5th Ave, Hines, IL 60141, USA
| | - Sonia Gonzalez
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, 2160 S 1st Avenue, Maywood, IL 60153, USA; Edward Hines Jr. VA Hospital, 5000 5th Ave, Hines, IL 60141, USA
| | - Yewon Rhee
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, 2160 S 1st Avenue, Maywood, IL 60153, USA; Edward Hines Jr. VA Hospital, 5000 5th Ave, Hines, IL 60141, USA
| | - Karie Scrogin
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, 2160 S 1st Avenue, Maywood, IL 60153, USA; Edward Hines Jr. VA Hospital, 5000 5th Ave, Hines, IL 60141, USA
| | - Rocco G Gogliotti
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, 2160 S 1st Avenue, Maywood, IL 60153, USA; Edward Hines Jr. VA Hospital, 5000 5th Ave, Hines, IL 60141, USA.
| |
Collapse
|
3
|
Li H, Liu S, Lin C, Wu Y, Wu X, Huang Y, Wu Y, Tong X, Xu X. Intravenous esketamine in pediatric Rett syndrome: An open-label, early phase 1 pilot study. Mol Ther Methods Clin Dev 2025; 33:101413. [PMID: 39990962 PMCID: PMC11847261 DOI: 10.1016/j.omtm.2025.101413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 01/16/2025] [Indexed: 02/25/2025]
Abstract
Rett syndrome (RTT) is a severe neurodevelopmental disorder. N-Methyl-d-aspartate receptor (NMDAR) antagonism has shown therapeutic potential in preclinical RTT models. We performed a pilot study to explore whether intravenous esketamine, an NMDAR antagonist, alleviates the symptoms of pediatric RTT. This was a prospective, single-arm, single-site, open-label, early phase 1 pilot study. Three girls with classic RTT aged 5-10 years were enrolled. Esketamine was intravenously administrated once per week for 5 weeks. The efficacy assessments included RTT-related questionnaires and video electroencephalograms (VEEGs). Prespecified adverse events (AEs) were monitored using clinical observations and standard laboratory tests. The treatment with intravenous esketamine was generally well tolerated and safe, with some patients experiencing mild AEs, including self-alleviating nausea, vomiting, and irritability. Three participants showed minimal improvements in their Clinical Global Impression Scale-Improvement, Rett Syndrome Behavior Questionnaire, and Revised Motor Behaviors Assessment Scale scores. However, individual differences were observed in the efficacy measures. VEEGs indicated gradual increases in posterior dominant rhythm peak frequency throughout the intervention. This pilot study highlights the potential of esketamine treatment for improving behavioral dysfunction in patients with RTT. Investigating the appropriate dosage form of esketamine may enhance its beneficial effects in RTT with fewer undesirable features.
Collapse
Affiliation(s)
- Huiping Li
- Department of Child Health Care, Xiamen Children’s Hospital, Children’s Hospital of Fudan University at Xiamen, Xiamen 361006, China
- Department of Child Health Care, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 201102, China
| | - Shu Liu
- Department of Child Health Care, Xiamen Children’s Hospital, Children’s Hospital of Fudan University at Xiamen, Xiamen 361006, China
| | - Caimei Lin
- Department of Neurology, Xiamen Children’s Hospital, Children’s Hospital of Fudan University at Xiamen, Xiamen 361006, China
| | - Yuchao Wu
- Department of Anesthesiology, Xiamen Children’s Hospital, Children’s Hospital of Fudan University at Xiamen, Xiamen 361006, China
| | - Xiuping Wu
- Pharmacy Department, Xiamen Children’s Hospital, Children’s Hospital of Fudan University at Xiamen, Xiamen 361006, China
| | - Yukun Huang
- Pediatric Intensive Care Unit, Xiamen Children’s Hospital, Children’s Hospital of Fudan University at Xiamen, Xiamen 361006, China
| | - Yajun Wu
- Department of Anesthesiology, Xiamen Children’s Hospital, Children’s Hospital of Fudan University at Xiamen, Xiamen 361006, China
| | - Xiubin Tong
- Department of Child Health Care, Xiamen Children’s Hospital, Children’s Hospital of Fudan University at Xiamen, Xiamen 361006, China
| | - Xiu Xu
- Department of Child Health Care, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 201102, China
| |
Collapse
|
4
|
Ährlund-Richter S, Harpe J, Fernandes G, Lam R, Sur M. Persistent Disruptions in Prefrontal Connectivity Despite Behavioral Rescue by Environmental Enrichment in a Mouse Model of Rett Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637474. [PMID: 39990439 PMCID: PMC11844379 DOI: 10.1101/2025.02.10.637474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Rett Syndrome, a neurodevelopmental disorder caused by loss-of-function mutations in the MECP2 gene, is characterized by severe motor, cognitive and emotional impairments. Some of the deficits may result from changes in cortical connections, especially downstream projections of the prefrontal cortex, which may also be targets of restoration following rearing conditions such as environmental enrichment that alleviate specific symptoms. Here, using a heterozygous Mecp2 +/- female mouse model closely analogous to human Rett Syndrome, we investigated the impact of early environmental enrichment on behavioral deficits and prefrontal cortex connectivity. Behavioral analyses revealed that enriched housing rescued fine motor deficits and reduced anxiety, with enrichment-housed Mecp2 +/- mice performing comparably to wild-type (WT) controls in rotarod and open field assays. Anatomical mapping of top-down anterior cingulate cortex (ACA) projections demonstrated altered prefrontal cortex connectivity in Mecp2 +/- mice, with increased axonal density in the somatosensory cortex and decreased density in the motor cortex compared to WT controls. ACA axons revealed shifts in hemispheric distribution, particularly in the medial network regions, with Mecp2 +/- mice exhibiting reduced ipsilateral dominance. These changes were unaffected by enriched housing, suggesting that structural abnormalities in prefrontal cortex connectivity persist despite behavioral improvements. Enriched housing rescued brain-derived neurotrophic factor (BDNF) levels in the hippocampus but failed to restore BDNF levels in the prefrontal cortex, consistent with the persistent deficits observed in prefrontal axonal projections. These findings highlight the focal nature of changes induced by reduction of MeCP2 and by exposure to environmental enrichment, and suggest that environmental enrichment starting in adolescence can alleviate behavioral deficits without reversing abnormalities in large-scale cortical connectivity.
Collapse
Affiliation(s)
- Sofie Ährlund-Richter
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jonathan Harpe
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Giselle Fernandes
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ruby Lam
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mriganka Sur
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
5
|
Campbell K, Neul JL, Lieberman DN, Berry-Kravis E, Benke TA, Fu C, Percy A, Suter B, Morris D, Carpenter RL, Marsh ED, von Hehn J. A randomized, placebo-controlled, cross-over trial of ketamine in Rett syndrome. J Neurodev Disord 2025; 17:4. [PMID: 39856538 PMCID: PMC11761732 DOI: 10.1186/s11689-025-09591-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Preclinical studies and anecdotal case reports support the potential therapeutic benefit of low-dose oral ketamine as a treatment of clinical symptoms in Rett syndrome (RTT); however, no controlled studies have been conducted in RTT to evaluate safety, tolerability and efficacy. DESIGN This was a sequentially initiated, dose-escalating cohort, placebo-controlled, double blind, randomized sequence, cross-over study of oral ketamine in 6-12-year-old girls with RTT to evaluate short-term safety and tolerability and explore efficacy. METHODS Participants were randomized to either five days treatment with oral ketamine or matched placebo, followed by a nine-day wash-out period and then crossed-over to the opposite treatment. Ketamine was dosed twice daily at 0.75 mg/kg/dose (Cohort 1) or 1.5 mg/kg/dose (Cohort 2). An independent safety monitoring committee evaluated safety and approved proceeding to the next dose cohort. Caregivers, participants, outcome assessors, and study staff except pharmacists were blinded to allocation. The primary endpoint was safety and tolerability. Exploratory efficacy endpoints included change in clinician- and caregiver-rated measures of RTT features, brain activity on electroencephalography, and wearable biosensors to measure respiration, heart rate, sleep, and activity. RESULTS Twenty-three participants enrolled (11 in Cohort 1, 12 in Cohort 2) from 3/12/2019-11/22/2021. One participant was excluded from analysis due to not meeting inclusion criteria on blinded review prior to analysis. One participant was withdrawn from the study due to an adverse event (vomiting) after the first dose of ketamine. Although planned for four dose cohorts, the trial was stopped after Cohort 2 due to enrollment challenges associated with the COVID-19 pandemic. Ketamine was safe and tolerated in both cohorts, with 1 related treatment emergent adverse event of vomiting. No difference was observed in efficacy between ketamine and placebo. Electroencephalography showed the expected increase in high frequency power with ketamine. CONCLUSIONS Short-term, low-dose oral ketamine was safe and well tolerated in girls with RTT. No clinical efficacy of ketamine in treating symptoms of RTT was observed with 5 days of treatment, despite electroencephalography evidence of ketamine target engagement during the first dose. Further studies are needed to evaluate safety and efficacy of higher dose and longer exposure to ketamine in RTT. TRIAL REGISTRATION Registered at clinicaltrials.gov NCT03633058.
Collapse
Affiliation(s)
- Kathleen Campbell
- Division of Developmental and Behavioral Pediatrics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jeffrey L Neul
- Department of Pediatrics, Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - David N Lieberman
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | | | - Tim A Benke
- Departments of Pediatrics, Pharmacology, Neurology and Otolaryngology, Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, CO, USA
| | - Cary Fu
- Department of Pediatrics, Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alan Percy
- School of Medicine, Department of Pediatrics, Neurology, Neurobiology, Genetics, and Psychology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Bernhard Suter
- Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | | | | | - Eric D Marsh
- Division of Neurology, Department of Neurology, The Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, USA.
| | | |
Collapse
|
6
|
Pepe G, Coco R, Corica D, Luppino G, Morabito LA, Lugarà C, Abbate T, Zirilli G, Aversa T, Stagi S, Wasniewska M. Endocrine disorders in Rett syndrome: a systematic review of the literature. Front Endocrinol (Lausanne) 2024; 15:1477227. [PMID: 39544232 PMCID: PMC11560452 DOI: 10.3389/fendo.2024.1477227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/15/2024] [Indexed: 11/17/2024] Open
Abstract
Background Rett syndrome (RTT) is an X-linked progressive neurodevelopmental disorder that involves mainly girls and is the second most frequent cause of genetic intellectual disability. RTT leads to neurological regression between 6 and 18 months of life and could be associated with a variable neurological impairment. However, RTT affects not only neurological function but also wide aspects of non-neurological organs. Recent data showed that the endocrine system is often involved in RTT patients, including disorders of growth, bone health, thyroid, puberty onset, and weight abnormalities However, systematic data on endocrinopathies in RTT are scarce and limited. Objective This review aims to analyze the prevalence and type of endocrine comorbidities in RTT population, to allow a precocious diagnosis and appropriate endocrinological management. Methods Systematic research was carried out from January 2000 to March 2024 through MEDLINE via PubMed, Scopus, and the Cochrane Library. Results After the selection phase, a total of 22 studies (1090 screened) met the inclusion criteria and were reported in the present review. Five studies were observational-retrospective, four were cross-sectional and case report or series, three were survey, prospective, and case-control, and finally one study for descriptive-transversal and longitudinal population-based study. The sample population consisted of multiethnic groups or single ethnic groups. The main endocrinopathies reported were malnutrition, bone alterations, and alterations of puberty onset. Conclusions Our analysis shows that endocrinopathies are not rare in RTT patients. Therefore, in the context of a multidisciplinary approach, accurate screening and monitoring for endocrinopathies should be recommended in all RTT patients, to improve clinical practice, healthcare management, and, finally, patients' quality of life.
Collapse
Affiliation(s)
- Giorgia Pepe
- Unit of Pediatrics, Department of Human Pathology of Adulthood and Childhood, University of Messina, Messina, Italy
| | - Roberto Coco
- Unit of Pediatrics, Department of Human Pathology of Adulthood and Childhood, University of Messina, Messina, Italy
| | - Domenico Corica
- Unit of Pediatrics, Department of Human Pathology of Adulthood and Childhood, University of Messina, Messina, Italy
| | - Giovanni Luppino
- Unit of Pediatrics, Department of Human Pathology of Adulthood and Childhood, University of Messina, Messina, Italy
| | - Letteria Anna Morabito
- Unit of Pediatrics, Department of Human Pathology of Adulthood and Childhood, University of Messina, Messina, Italy
| | - Cecilia Lugarà
- Unit of Pediatrics, Department of Human Pathology of Adulthood and Childhood, University of Messina, Messina, Italy
| | - Tiziana Abbate
- Unit of Pediatrics, Department of Human Pathology of Adulthood and Childhood, University of Messina, Messina, Italy
| | - Giuseppina Zirilli
- Unit of Pediatrics, Department of Human Pathology of Adulthood and Childhood, University of Messina, Messina, Italy
| | - Tommaso Aversa
- Unit of Pediatrics, Department of Human Pathology of Adulthood and Childhood, University of Messina, Messina, Italy
| | - Stefano Stagi
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Malgorzata Wasniewska
- Unit of Pediatrics, Department of Human Pathology of Adulthood and Childhood, University of Messina, Messina, Italy
| |
Collapse
|
7
|
Dong HW, Weiss K, Baugh K, Meadows MJ, Niswender CM, Neul JL. Potentiation of the muscarinic acetylcholine receptor 1 modulates neurophysiological features in a mouse model of Rett syndrome. Neurotherapeutics 2024; 21:e00384. [PMID: 38880672 PMCID: PMC11284553 DOI: 10.1016/j.neurot.2024.e00384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 06/18/2024] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder primarily caused by mutations in the X chromosome-linked gene Methyl-CpG Binding Protein 2 (MECP2). Restoring MeCP2 expression after disease onset in a mouse model of RTT reverses phenotypes, providing hope for development of treatments for RTT. Translatable biomarkers of improvement and treatment responses have the potential to accelerate both preclinical and clinical evaluation of targeted therapies in RTT. Studies in people with and mouse models of RTT have identified neurophysiological features, such as auditory event-related potentials, that correlate with disease severity, suggesting that they could be useful as biomarkers of disease improvement or early treatment response. We recently demonstrated that treatment of RTT mice with a positive allosteric modulator (PAM) of muscarinic acetylcholine subtype 1 receptor (M1) improved phenotypes, suggesting that modulation of M1 activity is a potential therapy in RTT. To evaluate whether neurophysiological features could be useful biomarkers to assess the effects of M1 PAM treatment, we acutely administered the M1 PAM VU0486846 (VU846) at doses of 1, 3, 10 and 30 mg/kg in wildtype and RTT mice. This resulted in an inverted U-shaped dose response with maximal improvement of AEP features at 3 mg/kg but with no marked effect on basal EEG power or epileptiform discharges in RTT mice and no significant changes in wildtype mice. These findings suggest that M1 potentiation can improve neural circuit synchrony to auditory stimuli in RTT mice and that neurophysiological features have potential as pharmacodynamic or treatment-responsive biomarkers for preclinical and clinical evaluation of putative therapies in RTT.
Collapse
Affiliation(s)
- Hong-Wei Dong
- Department of Pediatrics, Division of Neurology, Vanderbilt University Medical Center, USA; Vanderbilt University Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kelly Weiss
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, USA; Warren Center for Neuroscience Drug Discovery, School of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Kathryn Baugh
- Department of Pediatrics, Division of Neurology, Vanderbilt University Medical Center, USA
| | - Mac J Meadows
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, USA; Warren Center for Neuroscience Drug Discovery, School of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Colleen M Niswender
- Vanderbilt University Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, USA; Warren Center for Neuroscience Drug Discovery, School of Medicine, Vanderbilt University, Nashville, TN, USA; Vanderbilt Institute for Chemical Biology, Nashville, TN, USA; Vanderbilt Brain Institute, Nashville, TN, USA.
| | - Jeffrey L Neul
- Department of Pediatrics, Division of Neurology, Vanderbilt University Medical Center, USA; Vanderbilt University Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, USA; Vanderbilt Brain Institute, Nashville, TN, USA.
| |
Collapse
|
8
|
Hsu TT, Huang TN, Wang CY, Hsueh YP. Deep brain stimulation of the Tbr1-deficient mouse model of autism spectrum disorder at the basolateral amygdala alters amygdalar connectivity, whole-brain synchronization, and social behaviors. PLoS Biol 2024; 22:e3002646. [PMID: 39012916 PMCID: PMC11280143 DOI: 10.1371/journal.pbio.3002646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/26/2024] [Accepted: 06/21/2024] [Indexed: 07/18/2024] Open
Abstract
Autism spectrum disorders (ASDs) are considered neural dysconnectivity syndromes. To better understand ASD and uncover potential treatments, it is imperative to know and dissect the connectivity deficits under conditions of autism. Here, we apply a whole-brain immunostaining and quantification platform to demonstrate impaired structural and functional connectivity and aberrant whole-brain synchronization in a Tbr1+/- autism mouse model. We express a channelrhodopsin variant oChIEF fused with Citrine at the basolateral amygdala (BLA) to outline the axonal projections of BLA neurons. By activating the BLA under blue light theta-burst stimulation (TBS), we then evaluate the effect of BLA activation on C-FOS expression at a whole brain level to represent neural activity. We show that Tbr1 haploinsufficiency almost completely disrupts contralateral BLA axonal projections and results in mistargeting in both ipsilateral and contralateral hemispheres, thereby globally altering BLA functional connectivity. Based on correlated C-FOS expression among brain regions, we further show that Tbr1 deficiency severely disrupts whole-brain synchronization in the absence of salient stimulation. Tbr1+/- and wild-type (WT) mice exhibit opposing responses to TBS-induced amygdalar activation, reducing synchronization in WT mice but enhancing it in Tbr1+/- mice. Whole-brain modular organization and intermodule connectivity are also affected by Tbr1 deficiency and amygdalar activation. Following BLA activation by TBS, the synchronizations of the whole brain and the default mode network, a specific subnetwork highly relevant to ASD, are enhanced in Tbr1+/- mice, implying a potential ameliorating effect of amygdalar stimulation on brain function. Indeed, TBS-mediated BLA activation increases nose-to-nose social interactions of Tbr1+/- mice, strengthening evidence for the role of amygdalar connectivity in social behaviors. Our high-resolution analytical platform reveals the inter- and intrahemispheric connectopathies arising from ASD. Our study emphasizes the defective synchronization at a whole-brain scale caused by Tbr1 deficiency and implies a potential beneficial effect of deep brain stimulation at the amygdala for TBR1-linked autism.
Collapse
Affiliation(s)
- Tsan-Ting Hsu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Tzyy-Nan Huang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Chien-Yao Wang
- Institute of Information Science, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, Republic of China
| |
Collapse
|
9
|
Medeiros D, Ayala-Baylon K, Egido-Betancourt H, Miller E, Chapleau C, Robinson H, Phillips ML, Yang T, Longo FM, Li W, Pozzo-Miller L. A small-molecule TrkB ligand improves dendritic spine phenotypes and atypical behaviors in female Rett syndrome mice. Dis Model Mech 2024; 17:dmm050612. [PMID: 38785269 PMCID: PMC11139040 DOI: 10.1242/dmm.050612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/06/2024] [Indexed: 05/25/2024] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in MECP2, which encodes methyl-CpG-binding protein 2, a transcriptional regulator of many genes, including brain-derived neurotrophic factor (BDNF). BDNF levels are lower in multiple brain regions of Mecp2-deficient mice, and experimentally increasing BDNF levels improve atypical phenotypes in Mecp2 mutant mice. Due to the low blood-brain barrier permeability of BDNF itself, we tested the effects of LM22A-4, a brain-penetrant, small-molecule ligand of the BDNF receptor TrkB (encoded by Ntrk2), on dendritic spine density and form in hippocampal pyramidal neurons and on behavioral phenotypes in female Mecp2 heterozygous (HET) mice. A 4-week systemic treatment of Mecp2 HET mice with LM22A-4 restored spine volume in MeCP2-expressing neurons to wild-type (WT) levels, whereas spine volume in MeCP2-lacking neurons remained comparable to that in neurons from female WT mice. Female Mecp2 HET mice engaged in aggressive behaviors more than WT mice, the levels of which were reduced to WT levels by the 4-week LM22A-4 treatment. These data provide additional support to the potential usefulness of novel therapies not only for RTT but also to other BDNF-related disorders.
Collapse
Affiliation(s)
- Destynie Medeiros
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Karen Ayala-Baylon
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hailey Egido-Betancourt
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Eric Miller
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christopher Chapleau
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Holly Robinson
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Mary L. Phillips
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Tao Yang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Frank M. Longo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wei Li
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lucas Pozzo-Miller
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
10
|
Smith M, Dodis GE, Vanderplow AM, Gonzalez S, Rhee Y, Gogliotti RG. Potentiation of the M 1 muscarinic acetylcholine receptor normalizes neuronal activation patterns and improves apnea severity in Mecp2+/- mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.586099. [PMID: 38659804 PMCID: PMC11042204 DOI: 10.1101/2024.04.15.586099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder that is caused by loss-of-function mutations in the methyl-CpG binding protein 2 ( MeCP2 ) gene. RTT patients experience a myriad of debilitating symptoms, which include respiratory phenotypes that are often associated with lethality. Our previous work established that expression of the M 1 muscarinic acetylcholine receptor (mAchR) is decreased in RTT autopsy samples, and that potentiation of the M 1 receptor improves apneas in a mouse model of RTT; however, the population of neurons driving this rescue is unclear. Loss of Mecp2 correlates with excessive neuronal activity in cardiorespiratory nuclei. Since M 1 is found on cholinergic interneurons, we hypothesized that M 1 -potentiating compounds decrease apnea frequency by tempering brainstem hyperactivity. To test this, Mecp2 +/- and Mecp2 +/+ mice were screened for apneas before and after administration of the M 1 positive allosteric modulator (PAM) VU0453595 (VU595). Brains from the same mice were then imaged for c-Fos, ChAT, and Syto16 using whole-brain light-sheet microscopy to establish genotype and drug-dependent activation patterns that could be correlated with VU595's efficacy on apneas. The vehicle-treated Mecp2 +/- brain exhibited broad hyperactivity when coupled with the phenotypic prescreen, which was significantly decreased by administration of VU595, particularly in regions known to modulate the activity of respiratory nuclei (i.e. hippocampus and striatum). Further, the extent of apnea rescue in each mouse showed a significant positive correlation with c-Fos expression in non-cholinergic neurons in the striatum, thalamus, dentate gyrus, and within the cholinergic neurons of the brainstem. These results indicate that Mecp2 +/- mice are prone to hyperactivity in brain regions that regulate respiration, which can be normalized through M 1 potentiation.
Collapse
|
11
|
Pepe G, Coco R, Corica D, Di Rosa G, Bossowski F, Skorupska M, Aversa T, Stagi S, Wasniewska M. Prevalence of Endocrinopathies in a Cohort of Patients with Rett Syndrome: A Two-Center Observational Study. Genes (Basel) 2024; 15:287. [PMID: 38540345 PMCID: PMC10970698 DOI: 10.3390/genes15030287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 04/09/2024] Open
Abstract
Systematic data on endocrinopathies in Rett syndrome (RTT) patients remain limited and inconclusive. The aim of this retrospective observational two-center study was to assess the prevalence of endocrinopathies in a pediatric population of RTT patients. A total of 51 Caucasian patients (47 girls, 4 boys) with a genetically confirmed diagnosis of RTT were enrolled (mean age 9.65 ± 5.9 years). The patients were referred from the Rett Center of two Italian Hospitals for endocrinological evaluation. All the study population underwent clinical and auxological assessments and hormonal workups. MeCP2 mutations were detected in 38 cases (74.5%), CDKL5 deletions in 11 (21.6%), and FOXG1 mutations in 2 (3.9%). Overall, 40 patients were treated with anti-seizure medications. The most frequent endocrinological finding was short stature (47%), followed by menstrual cycle abnormalities (46.2%), weight disorders (45.1%), low bone mineral density (19.6%), hyperprolactinemia (13.7%) and thyroid disorders (9.8%). In the entire study population, endocrinopathies were significantly more frequent in patients with MeCP2 mutations (p = 0.0005), and epilepsy was more frequent in CDKL5 deletions (p = 0.02). In conclusion, our data highlighted that endocrinopathies are not rare in RTT, especially in patients with MeCP2 deletions. Therefore, in the context of a multidisciplinary approach, endocrinological evaluation should be recommended for RTT patients.
Collapse
Affiliation(s)
- Giorgia Pepe
- Pediatric Unit, Department of Human Pathology of Adulthood and Childhood, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (G.P.); (R.C.); (D.C.); (F.B.); (M.S.); (T.A.)
| | - Roberto Coco
- Pediatric Unit, Department of Human Pathology of Adulthood and Childhood, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (G.P.); (R.C.); (D.C.); (F.B.); (M.S.); (T.A.)
| | - Domenico Corica
- Pediatric Unit, Department of Human Pathology of Adulthood and Childhood, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (G.P.); (R.C.); (D.C.); (F.B.); (M.S.); (T.A.)
| | - Gabriella Di Rosa
- Child Neuropsychiatric Unit, Department of Human Pathology of Adulthood and Childhood, University of Messina, 98128 Messina, Italy;
| | - Filip Bossowski
- Pediatric Unit, Department of Human Pathology of Adulthood and Childhood, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (G.P.); (R.C.); (D.C.); (F.B.); (M.S.); (T.A.)
| | - Magdalena Skorupska
- Pediatric Unit, Department of Human Pathology of Adulthood and Childhood, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (G.P.); (R.C.); (D.C.); (F.B.); (M.S.); (T.A.)
| | - Tommaso Aversa
- Pediatric Unit, Department of Human Pathology of Adulthood and Childhood, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (G.P.); (R.C.); (D.C.); (F.B.); (M.S.); (T.A.)
| | - Stefano Stagi
- Department of Health Sciences, University of Florence, 50139 Florence, Italy;
- Meyer Children Hospital IRCCS, 50139 Florence, Italy
| | - Malgorzata Wasniewska
- Pediatric Unit, Department of Human Pathology of Adulthood and Childhood, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (G.P.); (R.C.); (D.C.); (F.B.); (M.S.); (T.A.)
| |
Collapse
|
12
|
Medeiros D, Ayala-Baylon K, Egido-Betancourt H, Miller E, Chapleau CA, Robinson HA, Phillips ML, Yang T, Longo F, Li W, Pozzo-Miller L. A small-molecule TrkB ligand improves dendritic spine phenotypes and atypical behaviors in female Rett syndrome mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.09.566435. [PMID: 37986936 PMCID: PMC10659425 DOI: 10.1101/2023.11.09.566435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in methyl-CpG-binding protein-2 (MECP2), encoding a transcriptional regulator of many genes, including brain-derived neurotrophic factor (Bdnf). BDNF mRNA and protein levels are lower in RTT autopsy brains and in multiple brain regions of Mecp2-deficient mice, and experimentally increasing BDNF levels improve atypical phenotypes in Mecp2 mutant mice. Due to the low blood-brain barrier permeability of BDNF itself, we tested the effects of a brain penetrant, small molecule ligand of its TrkB receptors. Applied in vitro, LM22A-4 increased dendritic spine density in pyramidal neurons in cultured hippocampal slices from postnatal day (P) 7 male Mecp2 knockout (KO) mice as much as recombinant BDNF, and both effects were prevented by the TrkB receptor inhibitors K-252a and ANA-12. Consistent with its partial agonist activity, LM22A-4 did not affect spine density in CA1 pyramidal neurons in slice cultures from male wildtype (WT) mice, where typical BDNF levels outcompete its binding to TrkB. To identify neurons of known genotypes in the "mosaic" brain of female Mecp2 heterozygous (HET) mice, we treated 4-6-month-old female MeCP2-GFP WT and HET mice with peripheral injections of LM22A-4 for 4 weeks. Surprisingly, mutant neurons lacking MeCP2-GFP showed dendritic spine volumes comparable to that in WT controls, while MeCP2-GFP-expressing neurons showed larger spines, similar to the phenotype we described in symptomatic male Mecp2 KO mice where all neurons lack MeCP2. Consistent with this non-cell-autonomous mechanism, a 4-week systemic treatment with LM22A-4 had an effect only in MeCP2-GFP-expressing neurons in female Mecp2 HET mice, bringing dendritic spine volumes down to WT control levels, and without affecting spines of MeCP2-GFP-lacking neurons. At the behavioral level, we found that female Mecp2 HET mice engaged in aggressive behaviors significantly more than WT controls, which were reduced to WT levels by a 4-week systemic treatment with LM22A-4. Altogether, these data revealed differences in dendritic spine size and altered behaviors in Mecp2 HET mice, while providing support to the potential usefulness of BDNF-related therapeutic approaches such as the partial TrkB agonist LM22A-4.
Collapse
|
13
|
Javed S, Chang YT, Cho Y, Lee YJ, Chang HC, Haque M, Lin YC, Huang WH. Smith-Magenis syndrome protein RAI1 regulates body weight homeostasis through hypothalamic BDNF-producing neurons and neurotrophin downstream signalling. eLife 2023; 12:RP90333. [PMID: 37956053 PMCID: PMC10642964 DOI: 10.7554/elife.90333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023] Open
Abstract
Retinoic acid-induced 1 (RAI1) haploinsufficiency causes Smith-Magenis syndrome (SMS), a genetic disorder with symptoms including hyperphagia, hyperlipidemia, severe obesity, and autism phenotypes. RAI1 is a transcriptional regulator with a pan-neural expression pattern and hundreds of downstream targets. The mechanisms linking neural Rai1 to body weight regulation remain unclear. Here we find that hypothalamic brain-derived neurotrophic factor (BDNF) and its downstream signalling are disrupted in SMS (Rai1+/-) mice. Selective Rai1 loss from all BDNF-producing cells or from BDNF-producing neurons in the paraventricular nucleus of the hypothalamus (PVH) induced obesity in mice. Electrophysiological recordings revealed that Rai1 ablation decreased the intrinsic excitability of PVHBDNF neurons. Chronic treatment of SMS mice with LM22A-4 engages neurotrophin downstream signalling and delayed obesity onset. This treatment also partially rescued disrupted lipid profiles, insulin intolerance, and stereotypical repetitive behaviour in SMS mice. These data argue that RAI1 regulates body weight and metabolic function through hypothalamic BDNF-producing neurons and that targeting neurotrophin downstream signalling might improve associated SMS phenotypes.
Collapse
Affiliation(s)
- Sehrish Javed
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill UniversityMontréalCanada
- Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health CentreMontréalCanada
| | - Ya-Ting Chang
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill UniversityMontréalCanada
- Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health CentreMontréalCanada
| | - Yoobin Cho
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill UniversityMontréalCanada
- Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health CentreMontréalCanada
| | - Yu-Ju Lee
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill UniversityMontréalCanada
- Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health CentreMontréalCanada
| | - Hao-Cheng Chang
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill UniversityMontréalCanada
- Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health CentreMontréalCanada
| | - Minza Haque
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill UniversityMontréalCanada
- Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health CentreMontréalCanada
| | - Yu Cheng Lin
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill UniversityMontréalCanada
- Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health CentreMontréalCanada
| | - Wei-Hsiang Huang
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill UniversityMontréalCanada
- Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health CentreMontréalCanada
| |
Collapse
|
14
|
Ramirez JM, Carroll MS, Burgraff N, Rand CM, Weese-Mayer DE. A narrative review of the mechanisms and consequences of intermittent hypoxia and the role of advanced analytic techniques in pediatric autonomic disorders. Clin Auton Res 2023; 33:287-300. [PMID: 37326924 DOI: 10.1007/s10286-023-00958-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 05/25/2023] [Indexed: 06/17/2023]
Abstract
Disorders of autonomic functions are typically characterized by disturbances in multiple organ systems. These disturbances are often comorbidities of common and rare diseases, such as epilepsy, sleep apnea, Rett syndrome, congenital heart disease or mitochondrial diseases. Characteristic of many autonomic disorders is the association with intermittent hypoxia and oxidative stress, which can cause or exaggerate a variety of other autonomic dysfunctions, making the treatment and management of these syndromes very complex. In this review we discuss the cellular mechanisms by which intermittent hypoxia can trigger a cascade of molecular, cellular and network events that result in the dysregulation of multiple organ systems. We also describe the importance of computational approaches, artificial intelligence and the analysis of big data to better characterize and recognize the interconnectedness of the various autonomic and non-autonomic symptoms. These techniques can lead to a better understanding of the progression of autonomic disorders, ultimately resulting in better care and management.
Collapse
Affiliation(s)
- Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA, 98101, USA.
- Departments of Neurological Surgery and Pediatrics, University of Washington School of Medicine, 1900 Ninth Avenue, Seattle, WA, 98101, USA.
| | - Michael S Carroll
- Data Analytics and Reporting, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Autonomic Medicine, Stanley Manne Children's Research Institute at Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Nicholas Burgraff
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA, 98101, USA
| | - Casey M Rand
- Division of Autonomic Medicine, Stanley Manne Children's Research Institute at Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Debra E Weese-Mayer
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Autonomic Medicine, Stanley Manne Children's Research Institute at Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| |
Collapse
|
15
|
Malar DS, Thitilertdecha P, Ruckvongacheep KS, Brimson S, Tencomnao T, Brimson JM. Targeting Sigma Receptors for the Treatment of Neurodegenerative and Neurodevelopmental Disorders. CNS Drugs 2023; 37:399-440. [PMID: 37166702 PMCID: PMC10173947 DOI: 10.1007/s40263-023-01007-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/18/2023] [Indexed: 05/12/2023]
Abstract
The sigma-1 receptor is a 223 amino acid-long protein with a recently identified structure. The sigma-2 receptor is a genetically unrelated protein with a similarly shaped binding pocket and acts to influence cellular activities similar to the sigma-1 receptor. Both proteins are highly expressed in neuronal tissues. As such, they have become targets for treating neurological diseases, including Alzheimer's disease (AD), Huntington's disease (HD), Parkinson's disease (PD), multiple sclerosis (MS), Rett syndrome (RS), developmental and epileptic encephalopathies (DEE), and motor neuron disease/amyotrophic lateral sclerosis (MND/ALS). In recent years, there have been many pre-clinical and clinical studies of sigma receptor (1 and 2) ligands for treating neurological disease. Drugs such as blarcamesine, dextromethorphan and pridopidine, which have sigma-1 receptor activity as part of their pharmacological profile, are effective in treating multiple aspects of several neurological diseases. Furthermore, several sigma-2 receptor ligands are under investigation, including CT1812, rivastigmine and SAS0132. This review aims to provide a current and up-to-date analysis of the current clinical and pre-clinical data of drugs with sigma receptor activities for treating neurological disease.
Collapse
Affiliation(s)
- Dicson S Malar
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Premrutai Thitilertdecha
- Siriraj Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kanokphorn S Ruckvongacheep
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Sirikalaya Brimson
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - James M Brimson
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand.
- Research, Innovation and International Affairs, Faculty of Allied Health Sciences, Chulalongkorn University, Room 409, ChulaPat-1 Building, 154 Rama 1 Road, Bangkok, 10330, Thailand.
| |
Collapse
|
16
|
Petazzi P, Jorge-Torres OC, Gomez A, Scognamiglio I, Serra-Musach J, Merkel A, Grases D, Xiol C, O’Callaghan M, Armstrong J, Esteller M, Guil S. Global Impairment of Immediate-Early Genes Expression in Rett Syndrome Models and Patients Linked to Myelination Defects. Int J Mol Sci 2023; 24:ijms24021453. [PMID: 36674969 PMCID: PMC9864472 DOI: 10.3390/ijms24021453] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 12/14/2022] [Accepted: 12/29/2022] [Indexed: 01/15/2023] Open
Abstract
Rett syndrome (RTT) is a severe neurodevelopmental disease caused almost exclusively by mutations to the MeCP2 gene. This disease may be regarded as a synaptopathy, with impairments affecting synaptic plasticity, inhibitory and excitatory transmission and network excitability. The complete understanding of the mechanisms behind how the transcription factor MeCP2 so profoundly affects the mammalian brain are yet to be determined. What is known, is that MeCP2 involvement in activity-dependent expression programs is a critical link between this protein and proper neuronal activity, which allows the correct maturation of connections in the brain. By using RNA-sequencing analysis, we found several immediate-early genes (IEGs, key mediators of activity-dependent responses) directly bound by MeCP2 at the chromatin level and upregulated in the hippocampus and prefrontal cortex of the Mecp2-KO mouse. Quantification of the IEGs response to stimulus both in vivo and in vitro detected an aberrant expression pattern in MeCP2-deficient neurons. Furthermore, altered IEGs levels were found in RTT patient's peripheral blood and brain regions of post-mortem samples, correlating with impaired expression of downstream myelination-related genes. Altogether, these data indicate that proper IEGs expression is crucial for correct synaptic development and that MeCP2 has a key role in the regulation of IEGs.
Collapse
Affiliation(s)
- Paolo Petazzi
- Josep Carreras Leukemia Research Institute, School of Medicine, University of Barcelona, Carrer Casanova 143, 400° floor, 08036 Barcelona, Spain
- RICORS-TERAV, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Olga Caridad Jorge-Torres
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, 08916 Barcelona, Spain
- Correspondence: (O.C.J.-T.); (S.G.); Tel.: +34-935572828 (O.C.J.-T. & S.G.)
| | - Antonio Gomez
- Biosciences Department, Faculty of Sciences and Technology (FCT), University of Vic-Central University of Catalonia (UVic-UCC), C. de la Laura, 13, 08500 Vic, Spain
| | - Iolanda Scognamiglio
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Jordi Serra-Musach
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Angelika Merkel
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, 08916 Barcelona, Spain
| | - Daniela Grases
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, 08916 Barcelona, Spain
| | - Clara Xiol
- Fundación San Juan de Dios, 08950 Barcelona, Spain
- Servei de Medicina Genètica i Molecular, Institut de Recerca Pediàtrica, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Mar O’Callaghan
- Clínica Rett, Neurology Department, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
- CIBER-ER (Biomedical Network Research Center for Rare Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Judith Armstrong
- Servei de Medicina Genètica i Molecular, Institut de Recerca Pediàtrica, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
- CIBER-ER (Biomedical Network Research Center for Rare Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Manel Esteller
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, 08916 Barcelona, Spain
- Centro de Investigacion Biomedica en Red Cancer (CIBERONC), 28029 Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), 08907 Barcelona, Spain
| | - Sonia Guil
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, 08916 Barcelona, Spain
- Germans Trias i Pujol Health Science Research Institute, Badalona, 08916 Barcelona, Spain
- Correspondence: (O.C.J.-T.); (S.G.); Tel.: +34-935572828 (O.C.J.-T. & S.G.)
| |
Collapse
|
17
|
Megagiannis P, Suresh R, Rouleau GA, Zhou Y. Reversibility and therapeutic development for neurodevelopmental disorders, insights from genetic animal models. Adv Drug Deliv Rev 2022; 191:114562. [PMID: 36183904 DOI: 10.1016/j.addr.2022.114562] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/30/2022] [Accepted: 09/24/2022] [Indexed: 01/24/2023]
Abstract
Neurodevelopmental Disorders (NDDs) encompass a broad spectrum of conditions resulting from atypical brain development. Over the past decades, we have had the fortune to witness enormous progress in diagnosis, etiology discovery, modeling, and mechanistic understanding of NDDs from both fundamental and clinical research. Here, we review recent neurobiological advances from experimental models of NDDs. We introduce several examples and highlight breakthroughs in reversal studies of phenotypes using genetically engineered models of NDDs. The in-depth understanding of brain pathophysiology underlying NDDs and evaluations of reversibility in animal models paves the foundation for discovering novel treatment options. We discuss how the expanding property of cutting-edge technologies, such as gene editing and AAV-mediated gene delivery, are leveraged in animal models for the therapeutic development of NDDs. We envision opportunities and challenges toward faithful modeling and fruitful clinical translation.
Collapse
Affiliation(s)
- Platon Megagiannis
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital; Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Rahul Suresh
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital; Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Guy A Rouleau
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital; Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Yang Zhou
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital; Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada.
| |
Collapse
|
18
|
Haase F, Singh R, Gloss B, Tam P, Gold W. Meta-Analysis Identifies BDNF and Novel Common Genes Differently Altered in Cross-Species Models of Rett Syndrome. Int J Mol Sci 2022; 23:11125. [PMID: 36232428 PMCID: PMC9570315 DOI: 10.3390/ijms231911125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/06/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Rett syndrome (RTT) is a rare disorder and one of the most abundant causes of intellectual disabilities in females. Single mutations in the gene coding for methyl-CpG-binding protein 2 (MeCP2) are responsible for the disorder. MeCP2 regulates gene expression as a transcriptional regulator as well as through epigenetic imprinting and chromatin condensation. Consequently, numerous biological pathways on multiple levels are influenced. However, the exact molecular pathways from genotype to phenotype are currently not fully elucidated. Treatment of RTT is purely symptomatic as no curative options for RTT have yet to reach the clinic. The paucity of this is mainly due to an incomplete understanding of the underlying pathophysiology of the disorder with no clinically useful common disease drivers, biomarkers, or therapeutic targets being identified. With the premise of identifying universal and robust disease drivers and therapeutic targets, here, we interrogated a range of RTT transcriptomic studies spanning different species, models, and MECP2 mutations. A meta-analysis using RNA sequencing data from brains of RTT mouse models, human post-mortem brain tissue, and patient-derived induced pluripotent stem cell (iPSC) neurons was performed using weighted gene correlation network analysis (WGCNA). This study identified a module of genes common to all datasets with the following ten hub genes driving the expression: ATRX, ADCY7, ADCY9, SOD1, CACNA1A, PLCG1, CCT5, RPS9, BDNF, and MECP2. Here, we discuss the potential benefits of these genes as therapeutic targets.
Collapse
Affiliation(s)
- Florencia Haase
- School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Kids Neuroscience Centre, Kids Research, Children’s Hospital at Westmead, Westmead, NSW 2145, Australia
- Molecular Neurobiology Research Laboratory, Kids Research, Children’s Hospital at Westmead, Westmead, NSW 2145, Australia
| | - Rachna Singh
- School of Medicine Sydney, The University of Notre Dame, Chippendale, NSW 2007, Australia
| | - Brian Gloss
- Westmead Research Hub, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia
| | - Patrick Tam
- School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Embryology Research Unit, Children’s Medical Research Institute, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Wendy Gold
- School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Kids Neuroscience Centre, Kids Research, Children’s Hospital at Westmead, Westmead, NSW 2145, Australia
- Molecular Neurobiology Research Laboratory, Kids Research, Children’s Hospital at Westmead, Westmead, NSW 2145, Australia
| |
Collapse
|
19
|
Horvath PM, Piazza MK, Kavalali ET, Monteggia LM. MeCP2 loss-of-function dysregulates microRNAs regionally and disrupts excitatory/inhibitory synaptic transmission balance. Hippocampus 2022; 32:610-623. [PMID: 35851733 PMCID: PMC9344394 DOI: 10.1002/hipo.23455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 06/11/2022] [Accepted: 06/25/2022] [Indexed: 11/06/2022]
Abstract
Rett syndrome is a leading cause of intellectual disability in females primarily caused by loss of function mutations in the transcriptional regulator MeCP2. Loss of MeCP2 leads to a host of synaptic phenotypes that are believed to underlie Rett syndrome pathophysiology. Synaptic deficits vary by brain region upon MeCP2 loss, suggesting distinct molecular alterations leading to disparate synaptic outcomes. In this study, we examined the contribution of MeCP2's newly described role in miRNA regulation to regional molecular and synaptic impairments. Two miRNAs, miR-101a and miR-203, were identified and confirmed as upregulated in MeCP2 KO mice in the hippocampus and cortex, respectively. miR-101a overexpression in hippocampal cultures led to opposing effects at excitatory and inhibitory synapses and in spontaneous and evoked neurotransmission, revealing the potential for a single miRNA to broadly regulate synapse function in the hippocampus. These results highlight the importance of regional alterations in miRNA expression and the specific impact on synaptic function with potential implications for Rett syndrome.
Collapse
Affiliation(s)
- Patricia M. Horvath
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA,Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Michelle K. Piazza
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA,Neuroscience Program, Vanderbilt University, Nashville, Tennessee, USA
| | - Ege T. Kavalali
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA,Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - Lisa M. Monteggia
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA,Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
20
|
Smith M, Arthur B, Cikowski J, Holt C, Gonzalez S, Fisher NM, Vermudez SAD, Lindsley CW, Niswender CM, Gogliotti RG. Clinical and Preclinical Evidence for M 1 Muscarinic Acetylcholine Receptor Potentiation as a Therapeutic Approach for Rett Syndrome. Neurotherapeutics 2022; 19:1340-1352. [PMID: 35670902 PMCID: PMC9587166 DOI: 10.1007/s13311-022-01254-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 12/04/2022] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder that is characterized by developmental regression, loss of communicative ability, stereotyped hand wringing, cognitive impairment, and central apneas, among many other symptoms. RTT is caused by loss-of-function mutations in a methyl-reader known as methyl-CpG-binding protein 2 (MeCP2), a protein that links epigenetic changes on DNA to larger chromatin structure. Historically, target identification for RTT has relied heavily on Mecp2 knockout mice; however, we recently adopted the alternative approach of performing transcriptional profiling in autopsy samples from RTT patients. Through this mechanism, we identified muscarinic acetylcholine receptors (mAChRs) as potential therapeutic targets. Here, we characterized a cohort of 40 temporal cortex samples from individuals with RTT and quantified significantly decreased levels of the M1, M2, M3, and M5 mAChRs subtypes relative to neurotypical controls. Of these four subtypes, M1 expression demonstrated a linear relationship with MeCP2 expression, such that M1 levels were only diminished in contexts where MeCP2 was also significantly decreased. Further, we show that M1 potentiation with the positive allosteric modulator (PAM) VU0453595 (VU595) rescued social preference, spatial memory, and associative memory deficits, as well as decreased apneas in Mecp2+/- mice. VU595's efficacy on apneas in Mecp2+/- mice was mediated by the facilitation of the transition from inspiration to expiration. Molecular analysis correlated rescue with normalized global gene expression patterns in the brainstem and hippocampus, as well as increased Gsk3β inhibition and NMDA receptor trafficking. Together, these data suggest that M1 PAMs could represent a new class of RTT therapeutics.
Collapse
Affiliation(s)
- Mackenzie Smith
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, IL, 60153, USA
- Edward Hines Jr. VA Hospital, Hines, IL, 60141, USA
| | - Bright Arthur
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Jakub Cikowski
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, IL, 60153, USA
- Edward Hines Jr. VA Hospital, Hines, IL, 60141, USA
| | - Calista Holt
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, IL, 60153, USA
- Edward Hines Jr. VA Hospital, Hines, IL, 60141, USA
| | - Sonia Gonzalez
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, IL, 60153, USA
- Edward Hines Jr. VA Hospital, Hines, IL, 60141, USA
| | - Nicole M Fisher
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Sheryl Anne D Vermudez
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37232, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA
| | - Rocco G Gogliotti
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, IL, 60153, USA.
- Edward Hines Jr. VA Hospital, Hines, IL, 60141, USA.
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
21
|
Chaudry S, Vasudevan N. mTOR-Dependent Spine Dynamics in Autism. Front Mol Neurosci 2022; 15:877609. [PMID: 35782388 PMCID: PMC9241970 DOI: 10.3389/fnmol.2022.877609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022] Open
Abstract
Autism Spectrum Conditions (ASC) are a group of neurodevelopmental disorders characterized by deficits in social communication and interaction as well as repetitive behaviors and restricted range of interests. ASC are complex genetic disorders with moderate to high heritability, and associated with atypical patterns of neural connectivity. Many of the genes implicated in ASC are involved in dendritic spine pruning and spine development, both of which can be mediated by the mammalian target of rapamycin (mTOR) signaling pathway. Consistent with this idea, human postmortem studies have shown increased spine density in ASC compared to controls suggesting that the balance between autophagy and spinogenesis is altered in ASC. However, murine models of ASC have shown inconsistent results for spine morphology, which may underlie functional connectivity. This review seeks to establish the relevance of changes in dendritic spines in ASC using data gathered from rodent models. Using a literature survey, we identify 20 genes that are linked to dendritic spine pruning or development in rodents that are also strongly implicated in ASC in humans. Furthermore, we show that all 20 genes are linked to the mTOR pathway and propose that the mTOR pathway regulating spine dynamics is a potential mechanism underlying the ASC signaling pathway in ASC. We show here that the direction of change in spine density was mostly correlated to the upstream positive or negative regulation of the mTOR pathway and most rodent models of mutant mTOR regulators show increases in immature spines, based on morphological analyses. We further explore the idea that these mutations in these genes result in aberrant social behavior in rodent models that is due to these altered spine dynamics. This review should therefore pave the way for further research on the specific genes outlined, their effect on spine morphology or density with an emphasis on understanding the functional role of these changes in ASC.
Collapse
|
22
|
Choi M, Ko SY, Seo JY, Kim DG, Lee H, Chung H, Son H. Autistic-like social deficits in hippocampal MeCP2 knockdown rat models are rescued by ketamine. BMB Rep 2022. [PMID: 35410641 PMCID: PMC9152577 DOI: 10.5483/bmbrep.2022.55.5.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Autism or autism spectrum disorder (ASD) is a behavioral syndrome characterized by persistent deficits in social interaction, and repetitive patterns of behavior, interests, or activities. The gene encoding Methyl-CpG binding protein 2 (MeCP2) is one of a few exceptional genes of established causal effect in ASD. Although genetically engineered mice studies may shed light on how MeCP2 loss affects synaptic activity patterns across the whole brain, such studies are not considered practical in ASD patients due to the overall level of impairment, and are technically challenging in mice. For the first time, we show that hippo-campal MeCP2 knockdown produces behavioral abnormalities associated with autism-like traits in rats, providing a new strategy to investigate the efficacy of therapeutics in ASD. Ketamine, an N-Methyl-D-aspartate (NMDA) blocker, has been proposed as a possible treatment for autism. Using the MeCP2 knockdown rats in conjunction with a rat model of valproic acid (VPA)-induced ASD, we examined gene expression and ASD behaviors upon ketamine treatment. We report that the core symptoms of autism in MeCP2 knockdown rats with social impairment recovered dramatically following a single treatment with ketamine.
Collapse
Affiliation(s)
- Miyeon Choi
- Hanyang Biomedical Research Institute, Hanyang University, Seoul 04763, Korea
| | - Seung Yeon Ko
- Hanyang Biomedical Research Institute, Hanyang University, Seoul 04763, Korea
| | - Jee Young Seo
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
| | - Do Gyeong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
| | - Huiju Lee
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
| | - Heekyoung Chung
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
| | - Hyeon Son
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
23
|
Tascini G, Dell'Isola GB, Mencaroni E, Di Cara G, Striano P, Verrotti A. Sleep Disorders in Rett Syndrome and Rett-Related Disorders: A Narrative Review. Front Neurol 2022; 13:817195. [PMID: 35299616 PMCID: PMC8923297 DOI: 10.3389/fneur.2022.817195] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/02/2022] [Indexed: 11/13/2022] Open
Abstract
Rett Syndrome (RTT) is a rare and severe X-linked developmental brain disorder that occurs primarily in females, with a ratio of 1:10.000. De novo mutations in the Methyl-CpG Binding protein 2 (MECP2) gene on the long arm of X chromosome are responsible for more than 95% cases of classical Rett. In the remaining cases (atypical Rett), other genes are involved such as the cyclin-dependent kinase-like 5 (CDKL5) and the forkhead box G1 (FOXG1). Duplications of the MECP2 locus cause MECP2 duplication syndrome (MDS) which concerns about 1% of male patients with intellectual disability. Sleep disorders are common in individuals with intellectual disability, while the prevalence in children is between 16 and 42%. Over 80% of individuals affected by RTT show sleep problems, with a higher prevalence in the first 7 years of life and some degree of variability in correlation to age and genotype. Abnormalities in circadian rhythm and loss of glutamate homeostasis play a key role in the development of these disorders. Sleep disorders, epilepsy, gastrointestinal problems characterize CDKL5 Deficiency Disorder (CDD). Sleep impairment is an area of overlap between RTT and MECP2 duplication syndrome along with epilepsy, regression and others. Sleep dysfunction and epilepsy are deeply linked. Sleep deprivation could be an aggravating factor of epilepsy and anti-comitial therapy could interfere in sleep structure. Epilepsy prevalence in atypical Rett syndrome with severe clinical phenotype is higher than in classical Rett syndrome. However, RTT present a significant lifetime risk of epilepsy too. Sleep disturbances impact on child's development and patients' families and the evidence for its management is still limited. The aim of this review is to analyze pathophysiology, clinical features, the impact on other comorbidities and the management of sleep disorders in Rett syndrome and Rett-related syndrome.
Collapse
Affiliation(s)
- Giorgia Tascini
- Department of Pediatrics, University of Perugia, Perugia, Italy
| | | | | | | | - Pasquale Striano
- Pediatric Neurology and Muscular Diseases Unit, IRCCS "G. Gaslini" Institute, Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | | |
Collapse
|
24
|
Li W. Excitation and Inhibition Imbalance in Rett Syndrome. Front Neurosci 2022; 16:825063. [PMID: 35250460 PMCID: PMC8894599 DOI: 10.3389/fnins.2022.825063] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
A loss of the excitation/inhibition (E/I) balance in the neural circuit has emerged as a common neuropathological feature in many neurodevelopmental disorders. Rett syndrome (RTT), a prevalent neurodevelopmental disorder that affects 1:10,000-15,000 women globally, is caused by loss-of-function mutations in the Methyl-CpG-binding Protein-2 (Mecp2) gene. E/I imbalance is recognized as the leading cellular and synaptic hallmark that is fundamental to diverse RTT neurological symptoms, including stereotypic hand movements, impaired motor coordination, breathing irregularities, seizures, and learning/memory dysfunctions. E/I balance in RTT is not homogeneously altered but demonstrates brain region and cell type specificity instead. In this review, I elaborate on the current understanding of the loss of E/I balance in a range of brain areas at molecular and cellular levels. I further describe how the underlying cellular mechanisms contribute to the disturbance of the proper E/I ratio. Last, I discuss current pharmacologic innervations for RTT and their role in modifying the E/I balance.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
25
|
Ramirez JM, Karlen-Amarante M, Wang JDJ, Huff A, Burgraff N. Breathing disturbances in Rett syndrome. HANDBOOK OF CLINICAL NEUROLOGY 2022; 189:139-151. [PMID: 36031301 PMCID: PMC10029146 DOI: 10.1016/b978-0-323-91532-8.00018-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Rett Syndrome is an X-linked neurological disorder characterized by behavioral and neurological regression, seizures, motor deficits, and dysautonomia. A particularly prominent presentation includes breathing abnormalities characterized by breathing irregularities, hyperventilation, repetitive breathholding during wakefulness, obstructive and central apneas during sleep, and abnormal responses to hypoxia and hypercapnia. The condition and pathology of the respiratory system is further complicated by dysfunctions of breathing-motor coordination, which is reflected in dysphagia. The discovery of the X-linked mutations in the MECP2 gene has transformed our understanding of the cellular and molecular mechanisms that are at the root of various clinical phenotypes. However, the genotype-phenotype relationship is complicated by various factors which include not only X-inactivation but also consequences of the intermittent hypoxia and oxidative stress associated with the breathing abnormalities.
Collapse
Affiliation(s)
- Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States; Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, United States.
| | - Marlusa Karlen-Amarante
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Jia-Der Ju Wang
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Alyssa Huff
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Nicholas Burgraff
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States
| |
Collapse
|
26
|
Collins BE, Neul JL. Rett Syndrome and MECP2 Duplication Syndrome: Disorders of MeCP2 Dosage. Neuropsychiatr Dis Treat 2022; 18:2813-2835. [PMID: 36471747 PMCID: PMC9719276 DOI: 10.2147/ndt.s371483] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/14/2022] [Indexed: 11/30/2022] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused predominantly by loss-of-function mutations in the gene Methyl-CpG-binding protein 2 (MECP2), which encodes the MeCP2 protein. RTT is a MECP2-related disorder, along with MECP2 duplication syndrome (MDS), caused by gain-of-function duplications of MECP2. Nearly two decades of research have advanced our knowledge of MeCP2 function in health and disease. The following review will discuss MeCP2 protein function and its dysregulation in the MECP2-related disorders RTT and MDS. This will include a discussion of the genetic underpinnings of these disorders, specifically how sporadic X-chromosome mutations arise and manifest in specific populations. We will then review current diagnostic guidelines and clinical manifestations of RTT and MDS. Next, we will delve into MeCP2 biology, describing the dual landscapes of methylated DNA and its reader MeCP2 across the neuronal genome as well as the function of MeCP2 as a transcriptional modulator. Following this, we will outline common MECP2 mutations and genotype-phenotype correlations in both diseases, with particular focus on mutations associated with relatively mild disease in RTT. We will also summarize decades of disease modeling and resulting molecular, synaptic, and behavioral phenotypes associated with RTT and MDS. Finally, we list several therapeutics in the development pipeline for RTT and MDS and available evidence of their safety and efficacy.
Collapse
Affiliation(s)
- Bridget E Collins
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
| | - Jeffrey L Neul
- Vanderbilt Kennedy Center, Departments of Pediatrics, Pharmacology, and Special Education, Vanderbilt University Medical Center and Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
27
|
Liang T, Wu F, Sun Y, Wang B. Electrophysiological Activity and Brain Network During Recovery of Propofol Anesthesia: A Stereoelectroencephalography-Based Analysis in Patients With Intractable Epilepsy—An Exploratory Research. Front Neurol 2021. [DOI: 10.3389/fneur.2021.694964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background: The oscillations and interactions between different brain areas during recovery of consciousness (ROC) from anesthesia in humans are poorly understood. Reliable stereoelectroencephalography (SEEG) signatures for transitions between unconsciousness and consciousness under anesthesia have not yet been fully identified.Objective: This study was designed to observe the change of electrophysiological activity during ROC and construct a ROC network based on SEEG data to describe the network property of cortical and deep areas during ROC from propofol-induced anesthetic epileptic patients.Methods: We analyzed SEEG data recorded from sixteen right-handed epileptic patients during ROC from propofol anesthesia from March 1, 2019, to December 31, 2019. Power spectrum density (PSD), correlation, and coherence were used to describe different brain areas' electrophysiological activity. The clustering coefficient, characteristic path length, modularity, network efficiency, degrees, and betweenness centrality were used to describe the network changes during ROC from propofol anesthesia. Statistical analysis was performed using MATLAB 2016b. The power spectral data from different contacts were analyzed using a one-way analysis of variance (ANOVA) test with Tukey's post-hoc correction. One sample t-test was used for the analysis of network property. Kolmogorov-Smirnov test was used to judge data distribution. Non-normal distribution was analyzed using the signed rank-sum test.Result: From the data of these 16 patients, 10 cortical, and 22 deep positions were observed. In this network, we observed that bilateral occipital areas are essential parts that have strong links with many regions. The recovery process is different in the bilateral cerebral cortex. Stage B (propofol 3.0-2.5 μg/ml) and E (propofol 1.5 μg/ml-ROC) play important roles during ROC exhibiting significant changes. The clustering coefficient gradually decreases with the recovery from anesthesia, and the changes mainly come from the cortical region. The characteristic path length and network efficiency do not change significantly during the recovery from anesthesia, and the changes of network modularity and clustering coefficient are similar. Deep areas tend to form functional modules. The left occipital lobe, the left temporal lobe, bilateral amygdala are essential nodes in the network. Some specific cortical regions (i.e., left angular gyrus, right angular gyrus, right temporal lobe, left temporal lobe, and right angular gyrus) and deep regions (i.e., right amygdala, left cingulate gyrus, right insular lobe, right amygdala) have more significant constraints on other regions.Conclusion: We verified that the bilateral cortex's recovery process is the opposite, which is not found in the deep regions. Significant PSD changes were observed in many areas at the beginning of stop infusion and near recovery. Our study found that during the ROC process, the modularity and clustering coefficient of the deep area network is significantly improved. However, the changes of the bilateral cerebral cortex were different. Power spectrum analysis shows that low-frequency EEG in anesthesia recovery accounts for a large proportion. The changes of the bilateral brain in the process of anesthesia recovery are different. The clustering coefficient gradually decreased with the recovery from anesthesia, and the changes mainly came from the cortical region. The characteristic path length and network efficiency do not change significantly during the recovery from anesthesia, and the changes of network modularity and clustering coefficient were similar. During ROC, the left occipital lobe, the left temporal lobe, bilateral amygdala were essential nodes in the network. The findings of the current study suggest SEEG as an effective tool for providing direct evidence of the anesthesia recovery mechanism.
Collapse
|
28
|
Wu Y, Cui N, Xing H, Zhong W, Arrowood C, Johnson CM, Jiang C. In vivo evidence for the cellular basis of central hypoventilation of Rett syndrome and pharmacological correction in the rat model. J Cell Physiol 2021; 236:8082-8098. [PMID: 34077559 DOI: 10.1002/jcp.30462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 04/13/2021] [Accepted: 05/08/2021] [Indexed: 12/29/2022]
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused mostly by mutations in the MECP2 gene. RTT patients show periodical hypoventilation attacks. The breathing disorder contributing to the high incidence of sudden death is thought to be due to depressed central inspiratory (I) activity via unknown cellular processes. Demonstration of such processes may lead to targets for pharmacological control of the RTT-type hypoventilation. We performed in vivo recordings from medullary respiratory neurons on the RTT rat model. To our surprise, both I and expiratory (E) neurons in the ventral respiratory column (VRC) increased their firing activity in Mecp2-null rats with severe hypoventilation. These I neurons including E-I phase-spanning and other I neurons remained active during apneas. Consistent with enhanced central I drive, ectopic phrenic discharges during expiration as well as apnea were observed in the Mecp2-null rats. Considering the increased I neuronal firing and ectopic phrenic activity, the RTT-type hypoventilation does not seem to be caused by depression in central I activity, neither reduced medullary I premotor output. This as well as excessive E neuronal firing as shown in our previous studies suggests inadequate synaptic inhibition for phase transition. We found that the abnormal respiratory neuronal firing, ectopic phrenic discharge as well as RTT-type hypoventilation all can be corrected by enhancing GABAergic inhibition. More strikingly, Mecp2-null rats reaching humane endpoints with severe hypoventilation can be rescued by GABAergic augmentation. Thus, defective GABAergic inhibition among respiratory neurons is likely to play a role in the RTT-type hypoventilation, which can be effectively controlled with pharmacological agents.
Collapse
Affiliation(s)
- Yang Wu
- Department of Biology, Georgia State University, Atlanta, Georgia, USA
| | - Ningren Cui
- Department of Biology, Georgia State University, Atlanta, Georgia, USA
| | - Hao Xing
- Department of Biology, Georgia State University, Atlanta, Georgia, USA
| | - Weiwei Zhong
- Department of Biology, Georgia State University, Atlanta, Georgia, USA
| | - Colin Arrowood
- Department of Biology, Georgia State University, Atlanta, Georgia, USA
| | | | - Chun Jiang
- Department of Biology, Georgia State University, Atlanta, Georgia, USA
| |
Collapse
|
29
|
Gigliucci V, Teutsch J, Woodbury-Smith M, Luoni M, Busnelli M, Chini B, Banerjee A. Region-Specific KCC2 Rescue by rhIGF-1 and Oxytocin in a Mouse Model of Rett Syndrome. Cereb Cortex 2021; 32:2885-2894. [PMID: 34791112 DOI: 10.1093/cercor/bhab388] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/17/2023] Open
Abstract
Rett syndrome (RTT) is characterized by dysfunction in neuronal excitation/inhibition (E/I) balance, potentially impacting seizure susceptibility via deficits in K+/Cl- cotransporter 2 (KCC2) function. Mice lacking the Methyl-CpG binding protein 2 (MeCP2) recapitulate many symptoms of RTT, and recombinant human insulin-like growth factor-1 (rhIGF-1) restores KCC2 expression and E/I balance in MeCP2 KO mice. However, clinical trial outcomes of rhIGF-1 in RTT have been variable, and increasing its therapeutic efficacy is highly desirable. To this end, the neuropeptide oxytocin (OXT) is promising, as it also critically modulates KCC2 function during early postnatal development. We measured basal KCC2 expression levels in MeCP2 KO mice and identified 3 key frontal brain regions showing KCC2 alterations in young adult mice, but not in postnatal P10 animals. We hypothesized that deficits in an IGF-1/OXT signaling crosstalk modulating KCC2 may occur in RTT during postnatal development. Consistently, we detected alterations of IGF-1 receptor and OXT receptor levels in those brain areas. rhIGF-1 and OXT treatments in KO mice rescued KCC2 expression in a region-specific and complementary manner. These results suggest that region-selective combinatorial pharmacotherapeutic strategies could be most effective at normalizing E/I balance in key brain regions subtending the RTT pathophysiology.
Collapse
Affiliation(s)
| | - Jasper Teutsch
- Neuroscience Theme, Biosciences Institute, Newcastle University, United Kingdom.,Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Marc Woodbury-Smith
- Neuroscience Theme, Biosciences Institute, Newcastle University, United Kingdom
| | - Mirko Luoni
- Stem Cells and Neurogenesis Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Marta Busnelli
- Institute of Neuroscience, CNR, Milan, Italy.,NeuroMi Milan Center for Neuroscience, Milan, Italy
| | - Bice Chini
- Institute of Neuroscience, CNR, Milan, Italy.,NeuroMi Milan Center for Neuroscience, Milan, Italy
| | - Abhishek Banerjee
- Neuroscience Theme, Biosciences Institute, Newcastle University, United Kingdom.,Brain Research Institute, University of Zurich, Zurich, Switzerland
| |
Collapse
|
30
|
Collapse of complexity of brain and body activity due to excessive inhibition and MeCP2 disruption. Proc Natl Acad Sci U S A 2021; 118:2106378118. [PMID: 34686597 DOI: 10.1073/pnas.2106378118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2021] [Indexed: 11/18/2022] Open
Abstract
Complex body movements require complex dynamics and coordination among neurons in motor cortex. Conversely, a long-standing theoretical notion supposes that if many neurons in motor cortex become excessively synchronized, they may lack the necessary complexity for healthy motor coding. However, direct experimental support for this idea is rare and underlying mechanisms are unclear. Here we recorded three-dimensional body movements and spiking activity of many single neurons in motor cortex of rats with enhanced synaptic inhibition and a transgenic rat model of Rett syndrome (RTT). For both cases, we found a collapse of complexity in the motor system. Reduced complexity was apparent in lower-dimensional, stereotyped brain-body interactions, neural synchrony, and simpler behavior. Our results demonstrate how imbalanced inhibition can cause excessive synchrony among movement-related neurons and, consequently, a stereotyped motor code. Excessive inhibition and synchrony may underlie abnormal motor function in RTT.
Collapse
|
31
|
High Behavioral Variability Mediated by Altered Neuronal Excitability in auts2 Mutant Zebrafish. eNeuro 2021; 8:ENEURO.0493-20.2021. [PMID: 34544758 PMCID: PMC8503961 DOI: 10.1523/eneuro.0493-20.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 01/28/2023] Open
Abstract
Autism spectrum disorders (ASDs) are characterized by abnormal behavioral traits arising from neural circuit dysfunction. While a number of genes have been implicated in ASDs, in most cases, a clear understanding of how mutations in these genes lead to circuit dysfunction and behavioral abnormality is absent. The autism susceptibility candidate 2 (AUTS2) gene is one such gene, associated with ASDs, intellectual disability and a range of other neurodevelopmental conditions. However, the role of AUTS2 in neural development and circuit function is not at all known. Here, we undertook functional analysis of Auts2a, the main homolog of AUTS2 in zebrafish, in the context of the escape behavior. Escape behavior in wild-type zebrafish is critical for survival and is therefore, reliable, rapid, and has well-defined kinematic properties. auts2a mutant zebrafish are viable, have normal gross morphology and can generate escape behavior with normal kinematics. However, the behavior is unreliable and delayed, with high trial-to-trial variability in the latency. Using calcium imaging we probed the activity of Mauthner neurons during otic vesicle (OV) stimulation and observed lower probability of activation and reduced calcium transients in the mutants. With direct activation of Mauthner by antidromic stimulation, the threshold for activation in mutants was higher than that in wild-type, even when inhibition was blocked. Taken together, these results point to reduced excitability of Mauthner neurons in auts2a mutant larvae leading to unreliable escape responses. Our results show a novel role for Auts2a in regulating neural excitability and reliability of behavior.
Collapse
|
32
|
Vermudez SAD, Gogliotti RG, Arthur B, Buch A, Morales C, Moxley Y, Rajpal H, Conn PJ, Niswender CM. Profiling beneficial and potential adverse effects of MeCP2 overexpression in a hypomorphic Rett syndrome mouse model. GENES, BRAIN, AND BEHAVIOR 2021; 21:e12752. [PMID: 34002468 PMCID: PMC8599502 DOI: 10.1111/gbb.12752] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/15/2021] [Accepted: 05/16/2021] [Indexed: 01/03/2023]
Abstract
De novo loss-of-function mutations in methyl-CpG-binding protein 2 (MeCP2) lead to the neurodevelopmental disorder Rett syndrome (RTT). Despite promising results from strategies aimed at increasing MeCP2 levels, additional studies exploring how hypomorphic MeCP2 mutations impact the therapeutic window are needed. Here, we investigated the consequences of genetically introducing a wild-type MECP2 transgene in the Mecp2 R133C mouse model of RTT. The MECP2 transgene reversed the majority of RTT-like phenotypes exhibited by male and female Mecp2 R133C mice. However, three core symptom domains were adversely affected in female Mecp2R133C/+ animals; these phenotypes resemble those observed in disease contexts of excess MeCP2. Parallel control experiments in Mecp2Null/+ mice linked these adverse effects to the hypomorphic R133C mutation. Collectively, these data provide evidence regarding the safety and efficacy of genetically overexpressing functional MeCP2 in Mecp2 R133C mice and suggest that personalized approaches may warrant consideration for the clinical assessment of MeCP2-targeted therapies.
Collapse
Affiliation(s)
- Sheryl Anne D. Vermudez
- Department of Pharmacology and Warren Center for Neuroscience Drug DiscoveryVanderbilt UniversityNashvilleTennesseeUSA
| | - Rocco G. Gogliotti
- Department of Molecular Pharmacology and NeuroscienceLoyola University ChicagoChicagoIllinoisUSA
| | - Bright Arthur
- Department of Pharmacology and Warren Center for Neuroscience Drug DiscoveryVanderbilt UniversityNashvilleTennesseeUSA
| | - Aditi Buch
- Department of Pharmacology and Warren Center for Neuroscience Drug DiscoveryVanderbilt UniversityNashvilleTennesseeUSA
| | - Clarissa Morales
- Department of Pharmacology and Warren Center for Neuroscience Drug DiscoveryVanderbilt UniversityNashvilleTennesseeUSA
| | - Yuta Moxley
- Department of Pharmacology and Warren Center for Neuroscience Drug DiscoveryVanderbilt UniversityNashvilleTennesseeUSA
| | - Hemangi Rajpal
- Department of Pharmacology and Warren Center for Neuroscience Drug DiscoveryVanderbilt UniversityNashvilleTennesseeUSA
| | - P. Jeffrey Conn
- Department of Pharmacology and Warren Center for Neuroscience Drug DiscoveryVanderbilt UniversityNashvilleTennesseeUSA,Vanderbilt Kennedy CenterVanderbilt UniversityNashvilleTennesseeUSA,Vanderbilt Institute of Chemical BiologyVanderbilt UniversityNashvilleTennesseeUSA
| | - Colleen M. Niswender
- Department of Pharmacology and Warren Center for Neuroscience Drug DiscoveryVanderbilt UniversityNashvilleTennesseeUSA,Vanderbilt Kennedy CenterVanderbilt UniversityNashvilleTennesseeUSA,Vanderbilt Institute of Chemical BiologyVanderbilt UniversityNashvilleTennesseeUSA
| |
Collapse
|
33
|
Scaramuzza L, De Rocco G, Desiato G, Cobolli Gigli C, Chiacchiaretta M, Mirabella F, Pozzi D, De Simone M, Conforti P, Pagani M, Benfenati F, Cesca F, Bedogni F, Landsberger N. The enhancement of activity rescues the establishment of Mecp2 null neuronal phenotypes. EMBO Mol Med 2021; 13:e12433. [PMID: 33665914 PMCID: PMC8033520 DOI: 10.15252/emmm.202012433] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 01/24/2021] [Accepted: 01/27/2021] [Indexed: 01/29/2023] Open
Abstract
MECP2 mutations cause Rett syndrome (RTT), a severe and progressive neurodevelopmental disorder mainly affecting females. Although RTT patients exhibit delayed onset of symptoms, several evidences demonstrate that MeCP2 deficiency alters early development of the brain. Indeed, during early maturation, Mecp2 null cortical neurons display widespread transcriptional changes, reduced activity, and defective morphology. It has been proposed that during brain development these elements are linked in a feed-forward cycle where neuronal activity drives transcriptional and morphological changes that further increase network maturity. We hypothesized that the enhancement of neuronal activity during early maturation might prevent the onset of RTT-typical molecular and cellular phenotypes. Accordingly, we show that the enhancement of excitability, obtained by adding to neuronal cultures Ampakine CX546, rescues transcription of several genes, neuronal morphology, and responsiveness to stimuli. Greater effects are achieved in response to earlier treatments. In vivo, short and early administration of CX546 to Mecp2 null mice prolongs lifespan, delays the disease progression, and rescues motor abilities and spatial memory, thus confirming the value for RTT of an early restoration of neuronal activity.
Collapse
Affiliation(s)
- Linda Scaramuzza
- Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
- Present address:
Department of Bioscience, University of Milan, Milan, Italy; Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”MilanItaly
| | - Giuseppina De Rocco
- Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
| | - Genni Desiato
- IRCCS Humanitas Research HospitalMilanItaly
- CNR Institute of NeuroscienceMilanItaly
| | - Clementina Cobolli Gigli
- Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
- Present address:
Francis Crick InstituteLondonUK
| | - Martina Chiacchiaretta
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di TecnologiaGenovaItaly
- Present address:
Department of NeuroscienceTufts University School of MedicineBostonMAUSA
| | - Filippo Mirabella
- IRCCS Humanitas Research HospitalMilanItaly
- Department of Biomedical Sciences, Humanitas UniversityMilanItaly
| | - Davide Pozzi
- IRCCS Humanitas Research HospitalMilanItaly
- Department of Biomedical Sciences, Humanitas UniversityMilanItaly
| | - Marco De Simone
- Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”MilanItaly
- Present address:
Department of Radiation Oncology, Cedars-Sinai Medical CenterLos Angeles, CAUSA
| | - Paola Conforti
- Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”MilanItaly
- Department of BiosciencesUniversity of MilanMilanItaly
| | - Massimiliano Pagani
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
- Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”MilanItaly
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di TecnologiaGenovaItaly
- IRCCS Ospedale Policlinico San MartinoGenovaItaly
- Present address:
Francis Crick InstituteLondonUK
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di TecnologiaGenovaItaly
- Department of Life SciencesUniversity of TriesteTriesteItaly
- Present address:
Department of Radiation Oncology, Cedars-Sinai Medical CenterLos Angeles, CAUSA
| | - Francesco Bedogni
- Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
- Present address:
Neuroscience and Mental Health Research Institute (NMHRI)Division of NeuroscienceSchool of BiosciencesCardiffUK
| | - Nicoletta Landsberger
- Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
| |
Collapse
|
34
|
Rett Syndrome: A Timely Review From Recognition to Current Clinical Approaches and Clinical Study Updates. Semin Pediatr Neurol 2021; 37:100881. [PMID: 33892852 DOI: 10.1016/j.spen.2021.100881] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/24/2021] [Accepted: 03/03/2021] [Indexed: 12/11/2022]
Abstract
Since the discovery of the genetic basis of Rett syndrome in 1999, our understanding has grown considerably both in the scientific and the clinical realms. In the last two decades, we have learned about the far-reaching effects of the aberrant MeCP2 protein, the growing list of involved genetic factors, and the genotype-phenotype clinical expression of common MECP2 mutations. This knowledge has led to several basic science research and clinical trials, focusing specifically on emerging treatments of Rett syndrome. As the pathophysiology behind the disease is better understood, treatments aimed at specific molecular targets will become available for clinicians to improve the life of individuals with Rett syndrome.
Collapse
|
35
|
Patrizi A, Awad PN, Chattopadhyaya B, Li C, Di Cristo G, Fagiolini M. Accelerated Hyper-Maturation of Parvalbumin Circuits in the Absence of MeCP2. Cereb Cortex 2021; 30:256-268. [PMID: 31038696 DOI: 10.1093/cercor/bhz085] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 02/21/2019] [Accepted: 03/29/2019] [Indexed: 12/27/2022] Open
Abstract
Methyl-CpG-binding protein 2 (MeCP2) mutations are the primary cause of Rett syndrome, a severe neurodevelopmental disorder. Cortical parvalbumin GABAergic interneurons (PV) make exuberant somatic connections onto pyramidal cells in the visual cortex of Mecp2-deficient mice, which contributes to silencing neuronal cortical circuits. This phenotype can be rescued independently of Mecp2 by environmental, pharmacological, and genetic manipulation. It remains unknown how Mecp2 mutation can result in abnormal inhibitory circuit refinement. In the present manuscript, we examined the development of GABAergic circuits in the primary visual cortex of Mecp2-deficient mice. We identified that PV circuits were the only GABAergic interneurons to be upregulated, while other interneurons were downregulated. Acceleration of PV cell maturation was accompanied by increased PV cells engulfment by perineuronal nets (PNNs) and by an increase of PV cellular and PNN structural complexity. Interestingly, selective deletion of Mecp2 from PV cells was sufficient to drive increased structure complexity of PNN. Moreover, the accelerated PV and PNN maturation was recapitulated in organotypic cultures. Our results identify a specific timeline of disruption of GABAergic circuits in the absence of Mecp2, indicating a possible cell-autonomous role of MeCP2 in the formation of PV cellular arbors and PNN structures in the visual cortex.
Collapse
Affiliation(s)
- Annarita Patrizi
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Schaller Research Group Leader at the German Cancer Research Center (DKFZ), Heildeberg, Germany
| | - Patricia N Awad
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Chloe Li
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Graziella Di Cristo
- Department of Neurosciences, Université de Montréal, Montreal, Canada.,CHU Ste Justine Research Center, Montreal, QC Canada
| | - Michela Fagiolini
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,International Research Center for Neurointelligence, University of Tokyo Institutes for Advanced Study, Tokyo, Japan
| |
Collapse
|
36
|
Grabrucker S, Pagano J, Schweizer J, Urrutia-Ruiz C, Schön M, Thome K, Ehret G, Grabrucker AM, Zhang R, Hengerer B, Bockmann J, Verpelli C, Sala C, Boeckers TM. Activation of the medial preoptic area (MPOA) ameliorates loss of maternal behavior in a Shank2 mouse model for autism. EMBO J 2021; 40:e104267. [PMID: 33491217 PMCID: PMC7917557 DOI: 10.15252/embj.2019104267] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 12/09/2020] [Accepted: 12/16/2020] [Indexed: 11/20/2022] Open
Abstract
Impairments in social relationships and awareness are features observed in autism spectrum disorders (ASDs). However, the underlying mechanisms remain poorly understood. Shank2 is a high‐confidence ASD candidate gene and localizes primarily to postsynaptic densities (PSDs) of excitatory synapses in the central nervous system (CNS). We show here that loss of Shank2 in mice leads to a lack of social attachment and bonding behavior towards pubs independent of hormonal, cognitive, or sensitive deficits. Shank2−/− mice display functional changes in nuclei of the social attachment circuit that were most prominent in the medial preoptic area (MPOA) of the hypothalamus. Selective enhancement of MPOA activity by DREADD technology re‐established social bonding behavior in Shank2−/− mice, providing evidence that the identified circuit might be crucial for explaining how social deficits in ASD can arise.
Collapse
Affiliation(s)
- Stefanie Grabrucker
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany.,Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - Jessica Pagano
- CNR Neuroscience Institute, Milan, Italy.,Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Johanna Schweizer
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | | | - Michael Schön
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Kevin Thome
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Günter Ehret
- Institute of Neurobiology, Ulm University, Ulm, Germany
| | - Andreas M Grabrucker
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Bernal Institute, University of Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland
| | - Rong Zhang
- Neuroscience Research Institute, Peking University, Beijing, China.,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China.,Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | | | - Jürgen Bockmann
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | | | - Carlo Sala
- CNR Neuroscience Institute, Milan, Italy
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany.,DZNE, Ulm Site, Ulm, Germany
| |
Collapse
|
37
|
Wong HHW, Rannio S, Jones V, Thomazeau A, Sjöström PJ. NMDA receptors in axons: there's no coincidence. J Physiol 2020; 599:367-387. [PMID: 33141440 DOI: 10.1113/jp280059] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 10/27/2020] [Indexed: 12/16/2022] Open
Abstract
In the textbook view, N-methyl-d-aspartate (NMDA) receptors are postsynaptically located detectors of coincident activity in Hebbian learning. However, controversial presynaptically located NMDA receptors (preNMDARs) have for decades been repeatedly reported in the literature. These preNMDARs have typically been implicated in the regulation of short-term and long-term plasticity, but precisely how they signal and what their functional roles are have been poorly understood. The functional roles of preNMDARs across several brain regions and different forms of plasticity can differ vastly, with recent discoveries showing key involvement of unusual subunit composition. Increasing evidence shows preNMDAR can signal through both ionotropic action by fluxing calcium and in metabotropic mode even in the presence of magnesium blockade. We argue that these unusual properties may explain why controversy has surrounded this receptor type. In addition, the expression of preNMDARs at some synapse types but not others can underlie synapse-type-specific plasticity. Last but not least, preNMDARs are emerging therapeutic targets in disease states such as neuropathic pain. We conclude that axonally located preNMDARs are required for specific purposes and do not end up there by accident.
Collapse
Affiliation(s)
- Hovy Ho-Wai Wong
- Department of Medicine, Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Ave, Montreal, Quebec, H3G 1A4, Canada
| | - Sabine Rannio
- Department of Medicine, Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Ave, Montreal, Quebec, H3G 1A4, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Victoria Jones
- Department of Medicine, Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Ave, Montreal, Quebec, H3G 1A4, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Aurore Thomazeau
- Department of Medicine, Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Ave, Montreal, Quebec, H3G 1A4, Canada
| | - P Jesper Sjöström
- Department of Medicine, Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Ave, Montreal, Quebec, H3G 1A4, Canada
| |
Collapse
|
38
|
Gomathi M, Padmapriya S, Balachandar V. Drug Studies on Rett Syndrome: From Bench to Bedside. J Autism Dev Disord 2020; 50:2740-2764. [PMID: 32016693 DOI: 10.1007/s10803-020-04381-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Drug studies on Rett syndrome (RTT) have drastically increased over the past few decades. This review aims to provide master data on bench-to-bedside drug studies involving RTT. A comprehensive literature review was performed by searching in PUBMED, MEDLINE and Google Scholar, international, national and regional clinical trial registries and pharmaceutical companies using the keywords "Rett syndrome treatment and/or drug or compound or molecule". Seventy drugs were investigated in non-clinical (N = 65 animal/cell line-based studies; N = 5 iPSC-based study) and clinical trials (N = 34) for ameliorating the symptoms of RTT. Though there is good progress in both clinical and non-clinical studies, none of these drugs entered phase III/IV for being launched as a therapeutic agent for RTT.
Collapse
Affiliation(s)
- Mohan Gomathi
- Human Molecular Genetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | | | - Vellingiri Balachandar
- Human Molecular Genetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India.
| |
Collapse
|
39
|
Ramirez JM, Karlen-Amarante M, Wang JDJ, Bush NE, Carroll MS, Weese-Mayer DE, Huff A. The Pathophysiology of Rett Syndrome With a Focus on Breathing Dysfunctions. Physiology (Bethesda) 2020; 35:375-390. [PMID: 33052774 PMCID: PMC7864239 DOI: 10.1152/physiol.00008.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/21/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023] Open
Abstract
Rett syndrome (RTT), an X-chromosome-linked neurological disorder, is characterized by serious pathophysiology, including breathing and feeding dysfunctions, and alteration of cardiorespiratory coupling, a consequence of multiple interrelated disturbances in the genetic and homeostatic regulation of central and peripheral neuronal networks, redox state, and control of inflammation. Characteristic breath-holds, obstructive sleep apnea, and aerophagia result in intermittent hypoxia, which, combined with mitochondrial dysfunction, causes oxidative stress-an important driver of the clinical presentation of RTT.
Collapse
Affiliation(s)
- Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, Washington
- Departments of Neurological Surgery and Pediatrics, University of Washington School of Medicine, Seattle, Washington
| | - Marlusa Karlen-Amarante
- Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, Washington
- Department of Physiology and Pathology, School of Dentistry of Araraquara, São Paulo State University (UNESP), Araraquara, Brazil
| | - Jia-Der Ju Wang
- Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, Washington
| | - Nicholas E Bush
- Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, Washington
| | - Michael S Carroll
- Data Analytics and Reporting, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of Autonomic Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Debra E Weese-Mayer
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of Autonomic Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Alyssa Huff
- Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
40
|
Ward CS, Huang TW, Herrera JA, Samaco RC, McGraw CM, Parra DE, Arvide EM, Ito-Ishida A, Meng X, Ure K, Zoghbi HY, Neul JL. Loss of MeCP2 Function Across Several Neuronal Populations Impairs Breathing Response to Acute Hypoxia. Front Neurol 2020; 11:593554. [PMID: 33193060 PMCID: PMC7662121 DOI: 10.3389/fneur.2020.593554] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/06/2020] [Indexed: 11/26/2022] Open
Abstract
Rett Syndrome (RTT) is a neurodevelopmental disorder caused by loss of function of the transcriptional regulator Methyl-CpG-Binding Protein 2 (MeCP2). In addition to the characteristic loss of hand function and spoken language after the first year of life, people with RTT also have a variety of physiological and autonomic abnormalities including disrupted breathing rhythms characterized by bouts of hyperventilation and an increased frequency of apnea. These breathing abnormalities, that likely involve alterations in both the circuitry underlying respiratory pace making and those underlying breathing response to environmental stimuli, may underlie the sudden unexpected death seen in a significant fraction of people with RTT. In fact, mice lacking MeCP2 function exhibit abnormal breathing rate response to acute hypoxia and maintain a persistently elevated breathing rate rather than showing typical hypoxic ventilatory decline that can be observed among their wild-type littermates. Using genetic and pharmacological tools to better understand the course of this abnormal hypoxic breathing rate response and the neurons driving it, we learned that the abnormal hypoxic breathing response is acquired as the animals mature, and that MeCP2 function is required within excitatory, inhibitory, and modulatory populations for a normal hypoxic breathing rate response. Furthermore, mice lacking MeCP2 exhibit decreased hypoxia-induced neuronal activity within the nucleus tractus solitarius of the dorsal medulla. Overall, these data provide insight into the neurons driving the circuit dysfunction that leads to breathing abnormalities upon loss of MeCP2. The discovery that combined dysfunction across multiple neuronal populations contributes to breathing dysfunction may provide insight into sudden unexpected death in RTT.
Collapse
Affiliation(s)
- Christopher S. Ward
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Teng-Wei Huang
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, United States
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, United States
| | - Jose A. Herrera
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, United States
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Rodney C. Samaco
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Christopher M. McGraw
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, United States
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, United States
| | - Diana E. Parra
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - E. Melissa Arvide
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Aya Ito-Ishida
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Xiangling Meng
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Kerstin Ure
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Huda Y. Zoghbi
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, United States
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, United States
| | - Jeffrey L. Neul
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, United States
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
41
|
Javed S, Selliah T, Lee YJ, Huang WH. Dosage-sensitive genes in autism spectrum disorders: From neurobiology to therapy. Neurosci Biobehav Rev 2020; 118:538-567. [PMID: 32858083 DOI: 10.1016/j.neubiorev.2020.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/26/2020] [Accepted: 08/17/2020] [Indexed: 12/24/2022]
Abstract
Autism spectrum disorders (ASDs) are a group of heterogenous neurodevelopmental disorders affecting 1 in 59 children. Syndromic ASDs are commonly associated with chromosomal rearrangements or dosage imbalance involving a single gene. Many of these genes are dosage-sensitive and regulate transcription, protein homeostasis, and synaptic function in the brain. Despite vastly different molecular perturbations, syndromic ASDs share core symptoms including social dysfunction and repetitive behavior. However, each ASD subtype has a unique pathogenic mechanism and combination of comorbidities that require individual attention. We have learned a great deal about how these dosage-sensitive genes control brain development and behaviors from genetically-engineered mice. Here we describe the clinical features of eight monogenic neurodevelopmental disorders caused by dosage imbalance of four genes, as well as recent advances in using genetic mouse models to understand their pathogenic mechanisms and develop intervention strategies. We propose that applying newly developed quantitative molecular and neuroscience technologies will advance our understanding of the unique neurobiology of each disorder and enable the development of personalized therapy.
Collapse
Affiliation(s)
- Sehrish Javed
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Tharushan Selliah
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Yu-Ju Lee
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Wei-Hsiang Huang
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.
| |
Collapse
|
42
|
Fagiolini M, Patrizi A, LeBlanc J, Jin LW, Maezawa I, Sinnett S, Gray SJ, Molholm S, Foxe JJ, Johnston MV, Naidu S, Blue M, Hossain A, Kadam S, Zhao X, Chang Q, Zhou Z, Zoghbi H. Intellectual and Developmental Disabilities Research Centers: A Multidisciplinary Approach to Understand the Pathogenesis of Methyl-CpG Binding Protein 2-related Disorders. Neuroscience 2020; 445:190-206. [PMID: 32360592 DOI: 10.1016/j.neuroscience.2020.04.037] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/20/2022]
Abstract
Disruptions in the gene encoding methyl-CpG binding protein 2 (MECP2) underlie complex neurodevelopmental disorders including Rett Syndrome (RTT), MECP2 duplication disorder, intellectual disabilities, and autism. Significant progress has been made on the molecular and cellular basis of MECP2-related disorders providing a new framework for understanding how altered epigenetic landscape can derail the formation and refinement of neuronal circuits in early postnatal life and proper neurological function. This review will summarize selected major findings from the past years and particularly highlight the integrated and multidisciplinary work done at eight NIH-funded Intellectual and Developmental Disabilities Research Centers (IDDRC) across the US. Finally, we will outline a path forward with identification of reliable biomarkers and outcome measures, longitudinal preclinical and clinical studies, reproducibility of results across centers as a synergistic effort to decode and treat the pathogenesis of the complex MeCP2 disorders.
Collapse
Affiliation(s)
- Michela Fagiolini
- Children's Hospital Intellectual and Developmental Disabilities Research Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Annarita Patrizi
- Children's Hospital Intellectual and Developmental Disabilities Research Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jocelyn LeBlanc
- Children's Hospital Intellectual and Developmental Disabilities Research Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lee-Way Jin
- UC Davis MIND Institute, University of California, Sacramento, CA, USA
| | - Izumi Maezawa
- UC Davis MIND Institute, University of California, Sacramento, CA, USA
| | - Sarah Sinnett
- UNC Intellectual and Developmental Disabilities Research Center, University of North Carolina, Gene Therapy Center and Dept. of Ophthalmology, Chapel Hill, NC, USA; Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Steven J Gray
- UNC Intellectual and Developmental Disabilities Research Center, University of North Carolina, Gene Therapy Center and Dept. of Ophthalmology, Chapel Hill, NC, USA; Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sophie Molholm
- The Cognitive Neurophysiology Laboratory, Departments of Pediatrics, Neuroscience, and Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - John J Foxe
- The Cognitive Neurophysiology Laboratory, Ernest J. Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Michael V Johnston
- Kennedy Krieger Institute Intellectual and Developmental Disabilities Research Center/Hugo Moser Research Institute at Kennedy Krieger and Johns Hopkins School of Medicine, USA
| | - Sakkubai Naidu
- Kennedy Krieger Institute Intellectual and Developmental Disabilities Research Center/Hugo Moser Research Institute at Kennedy Krieger and Johns Hopkins School of Medicine, USA
| | - Mary Blue
- Kennedy Krieger Institute Intellectual and Developmental Disabilities Research Center/Hugo Moser Research Institute at Kennedy Krieger and Johns Hopkins School of Medicine, USA
| | - Ahamed Hossain
- Kennedy Krieger Institute Intellectual and Developmental Disabilities Research Center/Hugo Moser Research Institute at Kennedy Krieger and Johns Hopkins School of Medicine, USA
| | - Shilpa Kadam
- Kennedy Krieger Institute Intellectual and Developmental Disabilities Research Center/Hugo Moser Research Institute at Kennedy Krieger and Johns Hopkins School of Medicine, USA
| | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Quiang Chang
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Zhaolan Zhou
- Department of Genetic, Epigenetic Institute, University of Pennsylvania Perelman School of Medicine, Intellectual and Developmental Disabilities Research Center, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Huda Zoghbi
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
43
|
Khoury ES, Sharma A, Ramireddy RR, Thomas AG, Alt J, Fowler A, Rais R, Tsukamoto T, Blue ME, Slusher B, Kannan S, Kannan RM. Dendrimer-conjugated glutaminase inhibitor selectively targets microglial glutaminase in a mouse model of Rett syndrome. Am J Cancer Res 2020; 10:5736-5748. [PMID: 32483415 PMCID: PMC7254984 DOI: 10.7150/thno.41714] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 04/14/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Elevated glutamate production and release from glial cells is a common feature of many CNS disorders. Inhibitors of glutaminase (GLS), the enzyme responsible for converting glutamine to glutamate have been developed to target glutamate overproduction. However, many GLS inhibitors have poor aqueous solubility, are unable to cross the blood brain barrier, or demonstrate significant toxicity when given systemically, precluding translation. Enhanced aqueous solubility and systemic therapy targeted to activated glia may address this challenge. Here we examine the impact of microglial-targeted GLS inhibition in a mouse model of Rett syndrome (RTT), a developmental disorder with no viable therapies, manifesting profound central nervous system effects, in which elevated glutamatergic tone, upregulation of microglial GLS, oxidative stress and neuroimmune dysregulation are key features. Methods: To enable this, we conjugated a potent glutaminase inhibitor, N-(5-{2-[2-(5-amino-[1,3,4]thiadiazol-2-yl)-ethylsulfanyl]-ethyl}-[1,3,4]thiadiazol-2-yl)-2-phenyl-acetamide (JHU29) to a generation 4 hydroxyl PAMAM dendrimer (D-JHU29). We then examined the effect of D-JHU29 in organotypic slice culture on glutamate release. We also examined GLS activity in microglial and non-microglial cells, and neurobehavioral phenotype after systemic administration of D-JHU29 in a mouse model of RTT. Results: We report successful conjugation of JHU29 to dendrimer resulting in enhanced water solubility compared to free JHU29. D-JHU29 reduced the excessive glutamate release observed in tissue culture slices in a clinically relevant Mecp2-knockout (KO) RTT mouse. Microglia isolated from Mecp2-KO mice demonstrated upregulation of GLS activity that normalized to wild-type levels following systemic treatment with D-JHU29. Neurobehavioral assessments in D-JHU29 treated Mecp2-KO mice revealed selective improvements in mobility. Conclusion: These findings demonstrate that glutaminase inhibitors conjugated to dendrimers are a viable mechanism to selectively inhibit microglial GLS to reduce glutamate production and improve mobility in a mouse model of RTT, with broader implications for selectively targeting this pathway in other neurodegenerative disorders.
Collapse
|
44
|
Piccolo FM, Liu Z, Dong P, Hsu CL, Stoyanova EI, Rao A, Tjian R, Heintz N. MeCP2 nuclear dynamics in live neurons results from low and high affinity chromatin interactions. eLife 2019; 8:51449. [PMID: 31868585 PMCID: PMC6957317 DOI: 10.7554/elife.51449] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/22/2019] [Indexed: 01/08/2023] Open
Abstract
Methyl-CpG-binding-Protein 2 (MeCP2) is an abundant nuclear protein highly enriched in neurons. Here we report live-cell single-molecule imaging studies of the kinetic features of mouse MeCP2 at high spatial-temporal resolution. MeCP2 displays dynamic features that are distinct from both highly mobile transcription factors and immobile histones. Stable binding of MeCP2 in living neurons requires its methyl-binding domain and is sensitive to DNA modification levels. Diffusion of unbound MeCP2 is strongly constrained by weak, transient interactions mediated primarily by its AT-hook domains, and varies with the level of chromatin compaction and cell type. These findings extend previous studies of the role of the MeCP2 MBD in high affinity DNA binding to living neurons, and identify a new role for its AT-hooks domains as critical determinants of its kinetic behavior. They suggest that limited nuclear diffusion of MeCP2 in live neurons contributes to its local impact on chromatin structure and gene expression.
Collapse
Affiliation(s)
- Francesco M Piccolo
- Laboratory of Molecular Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Zhe Liu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Peng Dong
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Ching-Lung Hsu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Elitsa I Stoyanova
- Laboratory of Molecular Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Anjana Rao
- La Jolla Institute for Allergy and Immunology, La Jolla, United States
| | - Robert Tjian
- Department of Molecular and Cell Biology, Li Ka Shing Center for Biomedical and Health Sciences, CIRM Center of Excellence, University of California, Howard Hughes Medical Institute, Berkeley, United States
| | - Nathaniel Heintz
- Laboratory of Molecular Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| |
Collapse
|
45
|
Smith ES, Smith DR, Eyring C, Braileanu M, Smith-Connor KS, Ei Tan Y, Fowler AY, Hoffman GE, Johnston MV, Kannan S, Blue ME. Altered trajectories of neurodevelopment and behavior in mouse models of Rett syndrome. Neurobiol Learn Mem 2019; 165:106962. [PMID: 30502397 PMCID: PMC8040058 DOI: 10.1016/j.nlm.2018.11.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 10/17/2018] [Accepted: 11/16/2018] [Indexed: 12/12/2022]
Abstract
Rett Syndrome (RTT) is a genetic disorder that is caused by mutations in the x-linked gene coding for methyl-CpG-biding-protein 2 (MECP2) and that mainly affects females. Male and female transgenic mouse models of RTT have been studied extensively, and we have learned a great deal regarding RTT neuropathology and how MeCP2 deficiency may be influencing brain function and maturation. In this manuscript we review what is known concerning structural and coinciding functional and behavioral deficits in RTT and in mouse models of MeCP2 deficiency. We also introduce our own corroborating data regarding behavioral phenotype and morphological alterations in volume of the cortex and striatum and the density of neurons, aberrations in experience-dependent plasticity within the barrel cortex and the impact of MeCP2 loss on glial structure. We conclude that regional structural changes in genetic models of RTT show great similarity to the alterations in brain structure of patients with RTT. These region-specific modifications often coincide with phenotype onset and contribute to larger issues of circuit connectivity, progression, and severity. Although the alterations seen in mouse models of RTT appear to be primarily due to cell-autonomous effects, there are also non-cell autonomous mechanisms including those caused by MeCP2-deficient glia that negatively impact healthy neuronal function. Collectively, this body of work has provided a solid foundation on which to continue to build our understanding of the role of MeCP2 on neuronal and glial structure and function, its greater impact on neural development, and potential new therapeutic avenues.
Collapse
Affiliation(s)
- Elizabeth S Smith
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Dani R Smith
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Charlotte Eyring
- The Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD 21205, USA
| | - Maria Braileanu
- Undergraduate Program in Neuroscience, The Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Karen S Smith-Connor
- The Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD 21205, USA
| | - Yew Ei Tan
- Perdana University Graduate School of Medicine, Kuala Lumpur, Malaysia
| | - Amanda Y Fowler
- Department of Biology, Morgan State University, Baltimore, MD 21251, USA
| | - Gloria E Hoffman
- Department of Biology, Morgan State University, Baltimore, MD 21251, USA
| | - Michael V Johnston
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD 21205, USA
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD 21205, USA
| | - Mary E Blue
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD 21205, USA.
| |
Collapse
|
46
|
Chowdhury T, Sternberg Z, Golanov E, Gelpi R, Rosemann T, Schaller BJ. Photic sneeze reflex: another variant of the trigeminocardiac reflex? FUTURE NEUROLOGY 2019. [DOI: 10.2217/fnl-2019-0007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The photic sneeze reflex (PSR) is a condition of uncontrollable sneezing episodes in response to bright light. This reflex often manifests as a mild phenomenon but may cause devastating consequences in some situations (aeroplane pilots, car drivers, etc.). Its exact mechanism is poorly understood. Interestingly, the roles of the fifth and tenth cranial nerves, brainstem nuclei and inciting patterns closely mimic a well-known brainstem reflex, known as the trigeminocardiac reflex (TCR). In this critical review, we hypothesize that the PSR can be a variant of the TCR. This concept will lead to a better understanding of the PSR and sharpens the TCR characteristics and open the doors for new research possibilities.
Collapse
Affiliation(s)
- Tumul Chowdhury
- Department of Anaesthesiology & Perioperative Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Zohara Sternberg
- Department of Neurology, Buffalo University of New York, NY, USA
| | - Eugene Golanov
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, USA & Weill Cornell Medicine, NY, USA
| | - Riccardo Gelpi
- Department of Cardiovascular Pathophysiology, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Thomas Rosemann
- Department of Primary Care, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
47
|
Kadam SD, Sullivan BJ, Goyal A, Blue ME, Smith-Hicks C. Rett Syndrome and CDKL5 Deficiency Disorder: From Bench to Clinic. Int J Mol Sci 2019; 20:ijms20205098. [PMID: 31618813 PMCID: PMC6834180 DOI: 10.3390/ijms20205098] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/08/2019] [Accepted: 10/11/2019] [Indexed: 12/18/2022] Open
Abstract
Rett syndrome (RTT) and CDKL5 deficiency disorder (CDD) are two rare X-linked developmental brain disorders with overlapping but distinct phenotypic features. This review examines the impact of loss of methyl-CpG-binding protein 2 (MeCP2) and cyclin-dependent kinase-like 5 (CDKL5) on clinical phenotype, deficits in synaptic- and circuit-homeostatic mechanisms, seizures, and sleep. In particular, we compare the overlapping and contrasting features between RTT and CDD in clinic and in preclinical studies. Finally, we discuss lessons learned from recent clinical trials while reviewing the findings from pre-clinical studies.
Collapse
Affiliation(s)
- Shilpa D Kadam
- The Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Brennan J Sullivan
- The Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA.
| | - Archita Goyal
- The Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA.
| | - Mary E Blue
- The Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Constance Smith-Hicks
- The Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
48
|
Banerjee A, Miller MT, Li K, Sur M, Kaufmann WE. Towards a better diagnosis and treatment of Rett syndrome: a model synaptic disorder. Brain 2019; 142:239-248. [PMID: 30649225 DOI: 10.1093/brain/awy323] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 10/31/2018] [Indexed: 12/21/2022] Open
Abstract
With the recent 50th anniversary of the first publication on Rett syndrome, and the almost 20 years since the first report on the link between Rett syndrome and MECP2 mutations, it is important to reflect on the tremendous advances in our understanding and their implications for the diagnosis and treatment of this neurodevelopmental disorder. Rett syndrome features an interesting challenge for biologists and clinicians, as the disorder lies at the intersection of molecular mechanisms of epigenetic regulation and neurophysiological alterations in synapses and circuits that together contribute to severe pathophysiological endophenotypes. Genetic, clinical, and neurobiological evidences support the notion that Rett syndrome is primarily a synaptic disorder, and a disease model for both intellectual disability and autism spectrum disorder. This review examines major developments in both recent neurobiological and preclinical findings of Rett syndrome, and to what extent they are beginning to impact our understanding and management of the disorder. It also discusses potential applications of knowledge on synaptic plasticity abnormalities in Rett syndrome to its diagnosis and treatment.
Collapse
Affiliation(s)
- Abhishek Banerjee
- Laboratory of Neural Circuit Dynamics, Brain Research Institute, University of Zürich, Zürich, Switzerland
| | - Meghan T Miller
- Roche Pharma Research and Early Development, Roche Innovation Center, F. Hoffman-La Roche, Basel, Switzerland
| | - Keji Li
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge MA, USA
| | - Mriganka Sur
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge MA, USA
| | - Walter E Kaufmann
- Department of Human Genetics, Emory University School of Medicine, Atlanta GA, USA
| |
Collapse
|
49
|
Walkley SU, Abbeduto L, Batshaw ML, Bhattacharyya A, Bookheimer SY, Christian BT, Constantino JN, de Vellis J, Doherty DA, Nelson DL, Piven J, Poduri A, Pomeroy SL, Samaco RC, Zoghbi HY, Guralnick MJ. Intellectual and developmental disabilities research centers: Fifty years of scientific accomplishments. Ann Neurol 2019; 86:332-343. [PMID: 31206741 PMCID: PMC8320680 DOI: 10.1002/ana.25531] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 06/12/2019] [Accepted: 06/13/2019] [Indexed: 12/17/2022]
Abstract
Progress in addressing the origins of intellectual and developmental disabilities accelerated with the establishment 50 years ago of the Eunice Kennedy Shriver National Institute of Child Health and Human Development of the National Institutes of Health and associated Intellectual and Developmental Disabilities Research Centers. Investigators at these Centers have made seminal contributions to understanding human brain and behavioral development and defining mechanisms and treatments of disorders of the developing brain. ANN NEUROL 2019;86:332-343.
Collapse
Affiliation(s)
- Steven U Walkley
- Department of Neuroscience, Albert Einstein College of Medicine, Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Bronx, NY
| | - Leonard Abbeduto
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, University of California, Davis Memory Impairments and Neurological Disorders Institute, Sacramento, CA
| | - Mark L Batshaw
- Children's Research Institute, Children's National Medical Center, Washington, DC
| | - Anita Bhattacharyya
- Department of Cell and Regenerative Biology, Waisman Center, University of Wisconsin-Madison, Madison, WI
| | - Susan Y Bookheimer
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Research Center, University of California, Los Angeles School of Medicine, Los Angeles, CA
| | - Bradley T Christian
- Departments of Medical Physics and Psychiatry, Waisman Center, University of Wisconsin-Madison, Madison, WI
| | - John N Constantino
- Departments of Psychiatry and Pediatrics, Washington University School of Medicine, Washington University in St Louis Intellectual and Developmental Disabilities Research Center, St Louis, MO
| | - Jean de Vellis
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Research Center, University of California, Los Angeles School of Medicine, Los Angeles, CA
| | - Daniel A Doherty
- Department of Pediatrics, Center on Human Development and Disability, University of Washington, Seattle, WA
| | - David L Nelson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Baylor College of Medicine Intellectual and Developmental Disabilities Research Center, Houston, TX
| | - Joseph Piven
- Carolina Institute for Developmental Disabilities, University of North Carolina, University of North Carolina Intellectual and Developmental Disabilities Research Center, Chapel Hill, NC
| | - Annapurna Poduri
- Department of Neurology, Harvard Medical School, Boston Children's Hospital and Harvard Medical School Intellectual and Developmental Disabilities Research Center, Boston, MA
| | - Scott L Pomeroy
- Department of Neurology, Harvard Medical School, Boston Children's Hospital and Harvard Medical School Intellectual and Developmental Disabilities Research Center, Boston, MA
| | - Rodney C Samaco
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Baylor College of Medicine Intellectual and Developmental Disabilities Research Center, Houston, TX
| | - Huda Y Zoghbi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Baylor College of Medicine Intellectual and Developmental Disabilities Research Center, Houston, TX
| | - Michael J Guralnick
- Departments of Psychology and Pediatrics, Center on Human Development and Disability, University of Washington, Seattle, WA
| |
Collapse
|
50
|
Abstract
Rett syndrome (RTT) is a severe neurological disorder caused by mutations in the gene encoding methyl-CpG-binding protein 2 (MeCP2). Almost two decades of research into RTT have greatly advanced our understanding of the function and regulation of the multifunctional protein MeCP2. Here, we review recent advances in understanding how loss of MeCP2 impacts different stages of brain development, discuss recent findings demonstrating the molecular role of MeCP2 as a transcriptional repressor, assess primary and secondary effects of MeCP2 loss and examine how loss of MeCP2 can result in an imbalance of neuronal excitation and inhibition at the circuit level along with dysregulation of activity-dependent mechanisms. These factors present challenges to the search for mechanism-based therapeutics for RTT and suggest specific approaches that may be more effective than others.
Collapse
|